Skip to main content
Erschienen in: BMC Cancer 1/2011

Open Access 01.12.2011 | Research article

A phenanthrene derived PARP inhibitor is an extra-centrosomes de-clustering agent exclusively eradicating human cancer cells

verfasst von: Asher Castiel, Leonid Visochek, Leonid Mittelman, Françoise Dantzer, Shai Izraeli, Malka Cohen-Armon

Erschienen in: BMC Cancer | Ausgabe 1/2011

Abstract

Background

Cells of most human cancers have supernumerary centrosomes. To enable an accurate chromosome segregation and cell division, these cells developed a yet unresolved molecular mechanism, clustering their extra centrosomes at two poles, thereby mimicking mitosis in normal cells. Failure of this bipolar centrosome clustering causes multipolar spindle structures and aberrant chromosomes segregation that prevent normal cell division and lead to 'mitotic catastrophe cell death'.

Methods

We used cell biology and biochemical methods, including flow cytometry, immunocytochemistry and live confocal imaging.

Results

We identified a phenanthrene derived PARP inhibitor, known for its activity in neuroprotection under stress conditions, which exclusively eradicated multi-centrosomal human cancer cells (mammary, colon, lung, pancreas, ovarian) while acting as extra-centrosomes de-clustering agent in mitosis. Normal human proliferating cells (endothelial, epithelial and mesenchymal cells) were not impaired. Despite acting as PARP inhibitor, the cytotoxic activity of this molecule in cancer cells was not attributed to PARP inhibition alone.

Conclusion

We identified a water soluble phenanthridine that exclusively targets the unique dependence of most human cancer cells on their supernumerary centrosomes bi-polar clustering for their survival. This paves the way for a new selective cancer-targeting therapy, efficient in a wide range of human cancers.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1471-2407-11-412) contains supplementary material, which is available to authorized users.
Asher Castiel, Leonid Visochek contributed equally to this work.

Competing interests

M. Cohen-Armon is the inventor of patent WO 2009/0477052, owned by the Tel-Aviv University. The co-authors declare that they have no competing interests. This study was not supported by any company or commercial fund.

Authors' contributions

LV and AC performed the experiments SI and AC contributed new analytical tools. LM performed the confocal measurements, FD-proofreading of the manuscript. MC-A designed the experiments and wrote the manuscript.
All authors read and approved the final manuscript.

Background

We have recently reported the selective eradication of human triple negative mammary cancer cells MDA-231 by phenanthrene derivatives (also acting as potent inhibitors of polyADP-ribose polymerases) [1]. These compounds included the phenanthridines PJ-34 and Phen and the isoquiniline Tiq-A. They were originally designed to protect neuronal cells in the central nervous system from cell death evoked by high activity of PARP-1 in response to DNA damage caused by brain injury, stroke or inflammation [2, 3]. We found that these molecules cause G2/M transition arrest in the cell cycle of both mammary cancer cells MDA-231 and normal epithelial cells MCF-10. However, while G2/M arrest was permanent in the cancer cells, and was accompanied by their massive cell death, normal mammary epithelial cells overcame the cell cycle arrest and continued to proliferate normally in the presence of these phenanthrene derivatives [1]. The most potent compound was PJ-34, which also efficiently prevented the development of MDA-231 xenotransplants in nude mice without inducing detectable toxic effects in the animals [1].
The current results outline a mechanism that apparently underlies the exclusive cytotoxicity of PJ-34 in these human mammary cancer cells. We found that this molecule acts as a centrosomes de-clustering agent in cells with supernumerary centrosomes, which are most abundant in these mammary cancer cells and in most human cancers [4, 5].
Bipolar centrosomes assembly during metaphase is crucial for bipolar spindle formation and accurate chromosomes segregation in cells undergoing mitosis [6, 7]. To accomplish these tasks, cells dividing with more than two centrosomes have developed a yet unresolved molecular mechanism, clustering their extra-centrosomes at two poles [4, 5, 8]. Failure of this bipolar centrosome assembly causes multipolar spindle structures and aberrant chromosomes segregation that prevent normal cell division [5]. This may lead either to 'mitotic slippage' where cells 'slip' out of mitosis to re-enter G1 without satisfying the spindle assembly checkpoint (SAC), or to induction of 'mitotic catastrophe cell death' [5, 9, 10] or 'anaphase catastrophy' [10]. Although cell death induced by failure to accomplish mitosis has been well documented, the underlying molecular mechanisms are still poorly understood [5, 10]
Extra centrosomes are most abundant in cells of human solid cancers and in some of the human hematological malignancies [4, 5], whereas in normal somatic cells the number of centrosomes is restricted, via several control mechanisms, to two centrosomes per cell [8, 9, 11]. This difference between normal and cancer human cells raised the idea of using compounds that interfere with the bipolar clustering of extra centrosomes to achieve selective eradication of cancer cells without harming normal tissues [5, 12].
Our findings confirm this hypothesis by identifying a molecule, which prevents extra centrosomes clustering in mitosis and exclusively eradicates human cancer cells with supernumerary centrosomes without impairing normal proliferating cells.

Methods

Cells and cell cultures

Human cancer cell lines included mammary triple negative (MDA-231) cells, lung (H1299), colon (DLD-1), ovarian (HeyA8), and pancreatic (Panc-1) cells. These cell-lines and the human epithelial cell line MCF-10A were supplied by ATCC, (Manassas, VA, USA). Human umbilical vein endothelial cells (Huvec) were supplied by PromoCell (Heidelberg, Germany), and human primary adipose-derived mesenchymal stroma cells were prepared from human thymus and kindly donated by Prof Jonathan Leor (Sheba Medical Center, Israel). The human cancer cell lines were chosen for our experiments as representatives of human cancer cells with high occurrence of extra-centrosomes.
Cancer cell lines were cultured in 6-well multi-dish plates (Nunc; Roskilde, Denmark) and maintained in a medium containing Dulbecco's Modified Eagle Medium (DMEM) containing 10% horse serum, 1% L-glutamine, and 1% Pen-Strep-Amphotericin B (Gibco, UK). Epithelial MCF-10A cells were cultured in DMEM/F12 (Gibco, UK) containing 6% fetal bovine serum (FBS), 0.02% epidermal growth factor (100 μg/ml; CytoLab, Rehovot, Israel), 2.8% hydrocortisone (50 μM; Sigma-Aldrich), 0.1% insulin (10 mg/ml; Sigma-Aldrich), and 1% Pen/Strep (Gibco, UK). Human endothelial cells, Huvec were cultured in endothelial cell growth medium (cat. # C22011; Promocell, Heidelberg, Germany) and used within 3 passages. Human primary mesenchymal stroma cells were prepared from thymus and cultured in DMEM/F12 (Rhenium) containing 15% fetal bovine serum (FBS). These cells were used within 5 passages.

Treatments

Cells were treated with phenanthrene-derived compounds acting as potent PARP inhibitors: PJ-34 (N-(6-oxo-5,6-dihydro-phenanthridin-2-yl)-N,N-dimethyl-acetamide), Tiq-A, (4H-thieno[2,3-c]isoquinolin-5-one), and Phen, (6(5H)-phenanthridinone). PJ-34 and Phen were purchased from Alexis Biochemicals (Enzo Life Sciences International, Inc. PA, USA), Tiq-A was purchased from Sigma-Aldrich. In addition, cells were treated with two potent PARP inhibitors that are not phenanthrene derivatives, ABT-888 (2-[(R)-2-methylpyrrolidin-2-yl]-1H-benzimidazole-4-carboxamide) (Abbott Labs, Illinois, U.S.A) and BSI-201 (4-iodo-3-nitrobenzamide) (BiPar, San Francisco, CA, USA) (Figure 1).
Flow cytometry was performed with Beckton Dickenson machine and the FlowJo software (Tree Star, Ashland, OR). It was used to monitor changes in the ploidy levels of malignant and normal cells. At the indicated times, the cells were collected, permeabilized (75% ethanol in double-distilled water), and stained with propidium iodide. Cell eradication and the kinetics of S-phase entry and G2/M transition were monitored by measuring the percentages of cells at the different phases. Cell counts at the selected time periods during 120 hours incubation indicated cell-cycle changes resulting from treatment with the tested compounds. Untreated cells were used as controls for each cell type at the selected time periods.

Confocal microscope imaging and immunocytochemistry

For immuno-cytochemistry, cells were grown on glass coverslips in the appropriate growth medium. After treatment, the cells were fixed with 4% paraformaldehyde (PFA; 20 minutes) and permeabilized with 0.2% Triton-X100 before blocking with 10% goat serum in PBS (details were described before [13]). The cells were immunolabeled for α and γ- tubulin. Spindles were labeled by antibodies directed against α-tubulin (T9026; Sigma) and detected by secondary fluorescent antibody Dylight (green) (Jackson ImmunoResearch Cat # 115-485-166). Antibodies directed against γ-tubulin (polyclonal, T5192; Sigma) detected by secondary fluorescent antibody CyTM3 (red) were used for immunolabeling of centrosomes. In some experiments centrosomes were double labeled for γ-tubulin (monoclonal, Abcam, MA, ab11316) and for centrin1 (polyclonal, Abcam, MA, ab11257) [14]. The percentage of multi-focal spindles was calculated in most cases out of a total number of 20 spindles that were detected in different randomly selected scanned samples of each of the tested cell types.
For live confocal imaging of cells, cells were transfected using liposomal reagent (Jet-PI, Polyplus, #101-10) with vectors encoding for γ-tubulin-GFP (for detection of centrosomes), α-tubulin-RFP (for detection of spindles and microtubule arrays) and for histone H2B-RED (for detection of chromosomes). Twenty-four hours after transfection the cells were exposed to PJ-34 (20 μM) for 18 hours, and then incubated in the presence of PJ-34 in live imaging chamber and scanned over-night (about 15 hours) by confocal microscope imaging. All the vectors were a gift of Prof. Michael Brandeis, Hebrew University, Jerusalem.
Luminescent detection of ATP level in the cells was used for quantification of cell viability. The Cell Viability Assay Kit (Abcam, MA) enabled a rapid quantification of viable cells in the cultures (the net result of growth arrest, cell death and cell proliferation). This method is based on the correlation between cell viability and ATP production, which signals the presence of metabolically active cells [15]. High percentage of cell death, as quantified by flow cytometry, was in correlation with decreased ATP levels in the tested cell cultures.

Results

We examined several potent PARP inhibitors, including the phenanthrene derivatives PJ-34, Tiq-A and Phen and the non-phenanthrene derivatives ABT-888 and BSI-201 (Figure 1) in human cancer cell types with a high occurrence of extra-centrosomal cells (>50%).
In our previous experiments PJ-34 caused G2/M arrest and a massive cell death in MDA-231 mammary cancer cells without impairing normal human mammary epithelial MCF-10 cells [1]. Given that MDA-231 cells have high occurrence of extra-centrosomes, and the interference with bipolar clustering of supernumerary centrosomes causes G2/M arrest [5, 16, 17], we examined the possibility that PJ-34 affects extra-centrosomes clustering in mitosis.
Our experiments disclosed a remarkable activity of PJ-34 on disruption of the bipolar clustering of supernumerary centrosomes in mammary MDA-231 cancer cells, causing distorted multi-polar spindles as early as 6 hours after PJ-34 application, followed by massive cell death within 48-72 hours (Figures 2a, b and Additional File 1). PJ-34 was applied only once to the cells, 24 hours after seeding, at twice to 3-times higher concentration than that completely inhibiting the activity of PARP-1 [3]. The cytotoxic activity of PJ-34 was identified in randomly selected fixed or live MDA-231 cells, scanned by confocal microscopy (Figures 2a, b and Additional File 1; Methods). Also, PJ-34 did not seem to impair the structure or formation of centrosomes, as indicated by centrosomes double labeling with centrin1 and γ-tubuline (Figure 2a) and by confocal imaging of centrosomes in live MDA-231 cells in the interphase (Additional File 2).
These findings associated the remarkable cytotoxic activity of PJ-34 in MDA-231 cells with de-clustering of their extra-centrosomes in mitosis, causing multi-polar distorted spindles and aberrant chromosomes segregation that lead to 'mitotic catastrophe cell death' [5].
Other phenanthrene derived molecules acting as PARP inhibitors, Phenanthridinon (Phen), and Tiq-A (Figure 1; [18, 19]), similarly interfered with centrosomes clustering in MDA-231 cells. However, their effect was milder and accompanied by a lower percentage of cell death (Figure 3).
In contrast, non-phenanthrene derivatives acting as potent PARP inhibitors, BSI-201 and ABT888 [18, 19], did not share this capability. Even when used at high concentrations (2-4 times higher than concentrations completely inhibiting the activity of PARP-1) they did not impair the bifocal clustering of extra centrosomes in these cells (Figure 3).
To further test the suggested contribution of PJ-34 to extra-centrosomes de-clustering, we examined its activity in other human cancer cell-lines with high occurrence of supernumerary centrosomes.
PJ-34 similarly eradicated the tested cells, including lung (H1299), colon (DLD-1), ovarian (HeyA8), and pancreatic (Panc-1) cancer cells. PJ-34 (20-30 μM) caused G2/M arrest in these cells and a subsequent massive cell death, observed 48-96 hours after PJ-34 application (Figures 4 and 5). In accordance, PJ-34 prevented the bipolar clustering of supernumerary centrosomes in these cells, causing multipolar spindles and aberrant chromosomes alignment detected within 18 hours after PJ-34 application (Figure 6). For the tested five types of cancer cell-lines, there was a significant correlation between the fraction of cells with multi-polar spindles and the reduction in cell viability (as indicated by the reduction in ATP level) of the treated cells (Figures 2a, 4 and 6; r = 0.92; p < 0.0001). Thus, cell death could be attributed to de-clustering of their extra-centrosomes.
In contrast, normal human proliferating cells were resistant to the cytotoxic activity of PJ-34, Tiq-A and Phen in human cancer cells. Even at the high concentrations used and the long incubation periods, the most potent compound, PJ-34, did not interfere with the cell cycle of benign human proliferating cells. The tested cells included human mammary epithelial cells MCF-10, mesenchymal cells prepared from human thymus and human umbilical vein endothelial cells (Huvec) (Figure 7). As for Huvec, PJ-34 did not affect their cell cycle within 48 hours incubation (Figure 7). These cells did not survive longer incubation periods even without treatment (> 5% cell death was measured in untreated Huvec cells after 72 hours incubation).
In these three types of benign cells, PJ-34 did not interfere with the formation and assembly of centrosomes (two centrosomes at the poles of bifocal spindles), nor with chromosomes segregation. Bipolar assembly of centrosomes, bifocal spindle formation and chromosomes segregation were not impaired in the normal proliferating cells, even after long incubation with high concentrations of PJ-34 (Figure 8).

Discussion

Our results suggest that the exclusive eradication of human cancer cells with extra centrosomes by the phenanthridine derivative PJ-34 is attributable to its extra-centrosomes de-clustering activity in mitosis (Figures 2, 4, 5, 6, Additional File 1 and Additional File 2).
According to our observations, PJ-34 did not impair centrosomes in both human normal and cancer cells, nor did it interfere with the formation of centrosomes in the interphase of multi-centrosomal cancer cells or impaired the bi-polar assembly of centrosomes in mitosis of normal proliferating cells with two centrosomes (Figures 2a, 8, Additional File 1 and Additional File 2). These features may underlie its outstanding capability to exclusively eradicate human cancer cells while keeping normal proliferating cells un-affected (Figures 5 and 7).
Theoretically, in cancer cells with high occurrence of extra-centrosomes (>50%) when mitosis is accompanied by eradication of multi-centrosomal cells, an exponential reduction of the fraction of bi-centrosomal cancer cells is expected. In this case, the higher their proliferation rate the more rapidly will these cancer cells be eradicated by extra-centrosomes de-clustering agents.
The indicated interference of the tested phenanthrene derivatives with the bipolar clustering of supernumerary centrosomes was not shared by non-phenanthrene derivatives acting as potent PARP inhibitors as well (Figure 3). This questions an exclusive role of PARP inhibition in extra centrosomes de-clustering. Nevertheless, these results could also reflect a possible, yet un-identified selective inhibition of PARP isoforms by phenanthridines.
Results obtained from genome-wide RNAi screen in multi-centrosomal cells, including embryonic Drosophila S2 cells and human malignant cells, identified genes required to suppress multipolar mitosis. Proteins involved in the organization and regulation of the cytoskeleton, including Kinesin HSET/Ncd (which is not required for polar organization in normal somatic cells) and the four chromosomal passenger complex (CPC) components, Aurora-B, INCENP, Survivin, and Borealin were identified among the main proteins required for bifocal clustering of extra-centrosomes in these cells [12, 17]. Evidence for the necessity of tankyrase-I and a putative human PARP-16 homolog in centrosomes clustering has been also provided [17, 20]. PolyADP-ribosylation of tankyrase-1 may contribute to spindle bipolarity by providing a static matrix, anchoring microtubule-associated motor proteins and spindle proteins [17, 20]. A specific role for PARP-16 in mitosis has not been identified yet [17].
PARP proteins are present in centrosomes [2124], and both Aurora-B and INCENP are targets for polyADP-ribosylation [22, 24]. In addition, supernumerary centrosomes were found in PARP-1 deficient cells [25]. Thus, in view of the exclusive interference of PJ-34 with extra centrosomes de-clustering in mitosis, a possible synergism between PARP activity and other mechanisms underlying extra-centrosomes clustering should be further investigated.
The indicated cytotoxic activity of the phenanthrene derivatives in human cancer cells with high occurrence of extra-centrosomes links for the first time between two well known, but poorly understood phenomena; Some anti-tumor effects of natural phenanthridines were first described in the early fifties [2628], and a higher cytotoxicity of some phenanthridines in cancer cells as compared to normal cells has been reported before [29, 30], although the underlying mechanisms have never been understood. In addition, cell-cycle arrest resulting from failure of centrosomes to accomplish bipolar clustering in multi-centrosomal cells has been acknowledged for nearly a century [6, 31, 32]. Nevertheless, the causal relationship between multiple centrosomes and malignancy has only recently been clarified [3235].
Mitotic-spindle microtubules are among the most effective targets for anti-cancer therapy [5, 10, 17, 3639]. Therefore efforts are invested in trying to find mechanisms exclusively targeting mitosis in cancer cells [4042].
Unlike the exclusive interference of PJ-34 with extra centrosomes bi-polar clustering in mitosis (Figures 2, 3, 4, 5, 6, Additional File 1 and Additional File 2), the currently known small molecules interfering with mitosis in cancer cells target microtubule polymerization (vinca alkaloids, taxanes, colchicines, and also griseofulvin). These compounds therefore impair normal cells as well, and cause severe side effects [3639].
The discovered potency of PJ-34 in preventing bipolar clustering of extra centrosomes in mitosis may pave the way for a new efficient therapy lacking intolerable side effects, which is based on compounds that exclusively target the unique dependence of many human cancer cells on their supernumerary centrosomes clustering for their survival.
Since many human cancer cells have high occurrence of multicentrosomal cells, the exclusive cytotoxic activity of PJ-34 in multi-centrosomal human cancer cells holds promise as a therapeutic tool for a selective chemotherapy in a wide range of human cancers, beyond the currently used selective therapeutic tools targeting specific genes/proteins that affect few cancer cell types [4345].

Conclusion

The identification of a molecule exclusively targeting the unique dependence of most human cancer cells on their supernumerary centrosomes bi-polar clustering for their survival, paves the way for a new highly selective cancer-targeting therapy, efficient in a wide range of human cancers.

Acknowledgements

This work was supported by the Feingrhot Karol and Leonora prize for cancer research (to M.C-A) and by the Israeli Science Foundation (to SI).
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

M. Cohen-Armon is the inventor of patent WO 2009/0477052, owned by the Tel-Aviv University. The co-authors declare that they have no competing interests. This study was not supported by any company or commercial fund.

Authors' contributions

LV and AC performed the experiments SI and AC contributed new analytical tools. LM performed the confocal measurements, FD-proofreading of the manuscript. MC-A designed the experiments and wrote the manuscript.
All authors read and approved the final manuscript.
Anhänge
Literatur
1.
Zurück zum Zitat Inbar-Rozensal D, Castiel A, Visochek L, Kastel D, Dantzer F, Izraeli S, Cohen-Armon M: A selective eradication of human nonhereditary breast cancer cells by phenanthridine-derived polyADP-ribose polymerase inhibitors. Breast Canc Res. 2009, 11/6/R78: 1-11. Inbar-Rozensal D, Castiel A, Visochek L, Kastel D, Dantzer F, Izraeli S, Cohen-Armon M: A selective eradication of human nonhereditary breast cancer cells by phenanthridine-derived polyADP-ribose polymerase inhibitors. Breast Canc Res. 2009, 11/6/R78: 1-11.
2.
Zurück zum Zitat Chiarugi A, Meli E, Calvani M, Picca R, Picca R, Baronti R, Camaioni E, et al: Novel isoquinolinone-derived inhibitors of polyADP-ribose polymerase-1: Pharmacological characterization and neuroprotective effects in an in-vitro model of cerebral ischemia. J Pharmacol Exp Ther. 2003, 305: 943-949. 10.1124/jpet.103.048934.CrossRefPubMed Chiarugi A, Meli E, Calvani M, Picca R, Picca R, Baronti R, Camaioni E, et al: Novel isoquinolinone-derived inhibitors of polyADP-ribose polymerase-1: Pharmacological characterization and neuroprotective effects in an in-vitro model of cerebral ischemia. J Pharmacol Exp Ther. 2003, 305: 943-949. 10.1124/jpet.103.048934.CrossRefPubMed
3.
Zurück zum Zitat Jagtap P, Szabo C: Poly(ADP-ribose)polymerase and the therapeutic effects of its inhibitors. Nat Rev Drug Discovery. 2005, 4: 421-440. 10.1038/nrd1718.CrossRefPubMed Jagtap P, Szabo C: Poly(ADP-ribose)polymerase and the therapeutic effects of its inhibitors. Nat Rev Drug Discovery. 2005, 4: 421-440. 10.1038/nrd1718.CrossRefPubMed
5.
Zurück zum Zitat Godinho SA, Kwon M, Pellman D: Centrosomes and cancer: how cancer cells divide with too many cetrosomes. Canc Met Rev. 2009, 28: 85-98. 10.1007/s10555-008-9163-6.CrossRef Godinho SA, Kwon M, Pellman D: Centrosomes and cancer: how cancer cells divide with too many cetrosomes. Canc Met Rev. 2009, 28: 85-98. 10.1007/s10555-008-9163-6.CrossRef
6.
Zurück zum Zitat Doxsey S: Re-evaluating centrosome function. Nat Rev Mol Cell Biol. 2001, 2: 688-698. 10.1038/35089575.CrossRefPubMed Doxsey S: Re-evaluating centrosome function. Nat Rev Mol Cell Biol. 2001, 2: 688-698. 10.1038/35089575.CrossRefPubMed
7.
Zurück zum Zitat Walczak CE, Heald R: Mechanisms of mitotic spindle assembly and function. International Rev of Cytology. 2008, (Elsevier Inc.), 265: 111-158. Walczak CE, Heald R: Mechanisms of mitotic spindle assembly and function. International Rev of Cytology. 2008, (Elsevier Inc.), 265: 111-158.
8.
Zurück zum Zitat Bettencourt-Dias M, Glover D: Centrosome biogenesis and function: centrosomics brings new understandings. Nat Rev Mol Cell Biol. 2007, 8 (6): 451-463. doi:10.1038/nrm2180.CrossRefPubMed Bettencourt-Dias M, Glover D: Centrosome biogenesis and function: centrosomics brings new understandings. Nat Rev Mol Cell Biol. 2007, 8 (6): 451-463. doi:10.1038/nrm2180.CrossRefPubMed
9.
Zurück zum Zitat Cunha-Ferreira I, Bento I, Bettencourt-Dias M: From zero to many: control of centriole number in development and disease. Traffic. 2009, 10: 482-498. 10.1111/j.1600-0854.2009.00905.x.CrossRefPubMed Cunha-Ferreira I, Bento I, Bettencourt-Dias M: From zero to many: control of centriole number in development and disease. Traffic. 2009, 10: 482-498. 10.1111/j.1600-0854.2009.00905.x.CrossRefPubMed
10.
Zurück zum Zitat Galimberti F, Thompson SL, Ravi S, Compton D, Dmitrovsky E: Anaphase Catastrophe Is a Target for Cancer Therapy. Clin Cancer Res. 2011, 17: 1218-1222. 10.1158/1078-0432.CCR-10-1178.CrossRefPubMedPubMedCentral Galimberti F, Thompson SL, Ravi S, Compton D, Dmitrovsky E: Anaphase Catastrophe Is a Target for Cancer Therapy. Clin Cancer Res. 2011, 17: 1218-1222. 10.1158/1078-0432.CCR-10-1178.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Tsou M-FB, Stearns T: Controlling centrosome number: licenses and blocks. Cur Opin Cell Biol. 2006, 18: 74-78. 10.1016/j.ceb.2005.12.008.CrossRef Tsou M-FB, Stearns T: Controlling centrosome number: licenses and blocks. Cur Opin Cell Biol. 2006, 18: 74-78. 10.1016/j.ceb.2005.12.008.CrossRef
12.
Zurück zum Zitat Leber B, Maier B, Fuchs F, Chi J, Riffel P, Anderhub S, Wagner L, Ho AD, Salisbury JL, Boutros M, Krämer A: Proteins Required for Centrosome Clustering in Cancer Cells. Sci Transl Med. 2010, 2 (33): DOI: 10.1126/scitranslmed.3000915. Leber B, Maier B, Fuchs F, Chi J, Riffel P, Anderhub S, Wagner L, Ho AD, Salisbury JL, Boutros M, Krämer A: Proteins Required for Centrosome Clustering in Cancer Cells. Sci Transl Med. 2010, 2 (33): DOI: 10.1126/scitranslmed.3000915.
13.
Zurück zum Zitat Cohen-Armon M, Visochek L, Rozensal D, Kalal A, Geistrikh I, Klein R, Bendetz-Nezer S, Yao Z, Seger R: DNA-independent PARP-1 activation by phosphorylated ERK2 increases Elk1 activity: a link to histone acetylation. Mol Cell. 2007, 25: 297-308. 10.1016/j.molcel.2006.12.012.CrossRefPubMed Cohen-Armon M, Visochek L, Rozensal D, Kalal A, Geistrikh I, Klein R, Bendetz-Nezer S, Yao Z, Seger R: DNA-independent PARP-1 activation by phosphorylated ERK2 increases Elk1 activity: a link to histone acetylation. Mol Cell. 2007, 25: 297-308. 10.1016/j.molcel.2006.12.012.CrossRefPubMed
14.
Zurück zum Zitat Stearns T, Evans L, Kirschner M: γ-Tubulin is a highly conserved component of the centrosome. Cell. 1991, 65: 825-836. 10.1016/0092-8674(91)90390-K.CrossRefPubMed Stearns T, Evans L, Kirschner M: γ-Tubulin is a highly conserved component of the centrosome. Cell. 1991, 65: 825-836. 10.1016/0092-8674(91)90390-K.CrossRefPubMed
15.
Zurück zum Zitat Vayssiere JL, Petit PX, Risler Y, Mignotte B: Commitment to apoptosis is associated with changes in mitochondrial biogenesis and activity in cell lines conditionally immortalized with simian virus 40. Proc Natl Acad Sci USA. 1994, 91: 11752-11756. 10.1073/pnas.91.24.11752.CrossRefPubMedPubMedCentral Vayssiere JL, Petit PX, Risler Y, Mignotte B: Commitment to apoptosis is associated with changes in mitochondrial biogenesis and activity in cell lines conditionally immortalized with simian virus 40. Proc Natl Acad Sci USA. 1994, 91: 11752-11756. 10.1073/pnas.91.24.11752.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Rieder CL, Maiato H: Stuck in Division or Passing through: What Happens When Cells Cannot Satisfy the Spindle Assembly Checkpoint. Dev Cell. 2004, 7: 637-651. 10.1016/j.devcel.2004.09.002.CrossRefPubMed Rieder CL, Maiato H: Stuck in Division or Passing through: What Happens When Cells Cannot Satisfy the Spindle Assembly Checkpoint. Dev Cell. 2004, 7: 637-651. 10.1016/j.devcel.2004.09.002.CrossRefPubMed
17.
Zurück zum Zitat Kwon M, Godinho SA, Chandhok NS, Ganem NJ, Azioune A, Thery M, Pellman D: Mechanisms to suppress multipolar divisions in cancer cells with extra centrosomes. Genes Dev. 2008, 22: 2189-2203. 10.1101/gad.1700908.CrossRefPubMedPubMedCentral Kwon M, Godinho SA, Chandhok NS, Ganem NJ, Azioune A, Thery M, Pellman D: Mechanisms to suppress multipolar divisions in cancer cells with extra centrosomes. Genes Dev. 2008, 22: 2189-2203. 10.1101/gad.1700908.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Rouleau M, Patel A, Hendze MJ, Kaufmann SH, Poirier GG: PARP inhibition: PARP1 and beyond. Nature Rev Cancer. 2010, 10: 293-301. 10.1038/nrc2812.CrossRef Rouleau M, Patel A, Hendze MJ, Kaufmann SH, Poirier GG: PARP inhibition: PARP1 and beyond. Nature Rev Cancer. 2010, 10: 293-301. 10.1038/nrc2812.CrossRef
19.
Zurück zum Zitat Krishnakumar R, Kraus WL: The PARP side of the nucleus: Molecular actions, physiological outcomes, and clinical targets. Mol Cell. 2010, 39: 8-24. 10.1016/j.molcel.2010.06.017.CrossRefPubMedPubMedCentral Krishnakumar R, Kraus WL: The PARP side of the nucleus: Molecular actions, physiological outcomes, and clinical targets. Mol Cell. 2010, 39: 8-24. 10.1016/j.molcel.2010.06.017.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Chang W, Dynek JN, Smith S: NuMA is a major acceptor of poly(ADP-ribosyl)ation by tankyrase 1 in mitosis. Biochem J. 2005, 391: 177-184. 10.1042/BJ20050885.CrossRefPubMedPubMedCentral Chang W, Dynek JN, Smith S: NuMA is a major acceptor of poly(ADP-ribosyl)ation by tankyrase 1 in mitosis. Biochem J. 2005, 391: 177-184. 10.1042/BJ20050885.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Chang P, Jacobson MK, Mitchison TJ: PolyADP-ribose is required for spindle assembly and structure. Nature. 2004, 432: 645-649. 10.1038/nature03061.CrossRefPubMed Chang P, Jacobson MK, Mitchison TJ: PolyADP-ribose is required for spindle assembly and structure. Nature. 2004, 432: 645-649. 10.1038/nature03061.CrossRefPubMed
22.
Zurück zum Zitat Earl E, Saxena A, MacDonald A, Hudson DF, Shaffer LG, et al: PolyADP-ribose polymers at active centromers and neocentromers at metaphase. Hum Mol Gen. 2000, 9: 187-194. 10.1093/hmg/9.2.187.CrossRef Earl E, Saxena A, MacDonald A, Hudson DF, Shaffer LG, et al: PolyADP-ribose polymers at active centromers and neocentromers at metaphase. Hum Mol Gen. 2000, 9: 187-194. 10.1093/hmg/9.2.187.CrossRef
23.
Zurück zum Zitat Saxena A, Saffery R, Wong LH, Kalitsis P, Choo A: Centromere Proteins Cenpa, Cenpb, and Bub3 Interact with Poly(ADP-ribose) Polymerase-1 Protein and Are Poly(ADP-ribosyl)ated. J Biol Chem. 2002, 277: 26921-26926. 10.1074/jbc.M200620200.CrossRefPubMed Saxena A, Saffery R, Wong LH, Kalitsis P, Choo A: Centromere Proteins Cenpa, Cenpb, and Bub3 Interact with Poly(ADP-ribose) Polymerase-1 Protein and Are Poly(ADP-ribosyl)ated. J Biol Chem. 2002, 277: 26921-26926. 10.1074/jbc.M200620200.CrossRefPubMed
24.
Zurück zum Zitat Kanai M, Tong WM, Sugihara E, Wang ZQ, Fukasawa K, Miwa1 M: Involvement of Poly(ADP-Ribose) Polymerase 1 and Poly(ADP-Ribosyl)ation in Regulation of Centrosome function. Mol Cell Biol. 2003, 23: 2451-2462. 10.1128/MCB.23.7.2451-2462.2003.CrossRefPubMedPubMedCentral Kanai M, Tong WM, Sugihara E, Wang ZQ, Fukasawa K, Miwa1 M: Involvement of Poly(ADP-Ribose) Polymerase 1 and Poly(ADP-Ribosyl)ation in Regulation of Centrosome function. Mol Cell Biol. 2003, 23: 2451-2462. 10.1128/MCB.23.7.2451-2462.2003.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Ogino H, Nozaki T, Gunji A, Maeda M, Suzuki H, Ohta T, Murakami Y, Nakagama H, Sugimura T, Masutani M: Loss of Parp-1 affects gene expression profile in a genome-wide manner in ES cells and liver cells. BMC Genomics. 2007, 8: 41-10.1186/1471-2164-8-41.CrossRefPubMedPubMedCentral Ogino H, Nozaki T, Gunji A, Maeda M, Suzuki H, Ohta T, Murakami Y, Nakagama H, Sugimura T, Masutani M: Loss of Parp-1 affects gene expression profile in a genome-wide manner in ES cells and liver cells. BMC Genomics. 2007, 8: 41-10.1186/1471-2164-8-41.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Coombs MM, Bhatt TS, Croft CJ: Correlation between carcinogenicity and chemical structure in Cyclopenta[α]- phenanthridines. Canc Res. 1973, 33: 832-837. Coombs MM, Bhatt TS, Croft CJ: Correlation between carcinogenicity and chemical structure in Cyclopenta[α]- phenanthridines. Canc Res. 1973, 33: 832-837.
27.
Zurück zum Zitat Phillips SD, Castle RN: A review of the chemistry of the antitumor benzo-[c]phenanthridine alkaloids nitidine and fagaronine and of the related antitumor alkaloid coralyne. Journal of Heterocyclic Chemistry. 1981, 18: 223-232. 10.1002/jhet.5570180202.CrossRef Phillips SD, Castle RN: A review of the chemistry of the antitumor benzo-[c]phenanthridine alkaloids nitidine and fagaronine and of the related antitumor alkaloid coralyne. Journal of Heterocyclic Chemistry. 1981, 18: 223-232. 10.1002/jhet.5570180202.CrossRef
28.
Zurück zum Zitat Turchi G, Glatt HR, Seidel A, Puliti A, Sbrana I: Structure activity relationship in the induction of chromosomal aberrations and spindle disturbances in Chinese hamster epithelial liver cells by regioisomeric phenanthridine quinines. Cell Biology and Toxicology. 1997, 13: 155-165. 10.1023/A:1007302111081.CrossRefPubMed Turchi G, Glatt HR, Seidel A, Puliti A, Sbrana I: Structure activity relationship in the induction of chromosomal aberrations and spindle disturbances in Chinese hamster epithelial liver cells by regioisomeric phenanthridine quinines. Cell Biology and Toxicology. 1997, 13: 155-165. 10.1023/A:1007302111081.CrossRefPubMed
29.
Zurück zum Zitat Lamoral-Theys D, Andolfi A, Van Goietsenoven G, Cimmino A, Le Calve B, Wauthoz N: Lycorine, the main phenanthridine Amaryllidaceae alkaloid, exhibits significant antitumor activity in cancer cells that display resistance to proapoptotic stimuli: an investigation of structure-activity relationship and mechanistic insight. J Med Chem. 2009, 52: 6244-6256. 10.1021/jm901031h.CrossRefPubMedPubMedCentral Lamoral-Theys D, Andolfi A, Van Goietsenoven G, Cimmino A, Le Calve B, Wauthoz N: Lycorine, the main phenanthridine Amaryllidaceae alkaloid, exhibits significant antitumor activity in cancer cells that display resistance to proapoptotic stimuli: an investigation of structure-activity relationship and mechanistic insight. J Med Chem. 2009, 52: 6244-6256. 10.1021/jm901031h.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Iwasaki H, Okabe T, Takara K, Toda T, Shimatani M, Oku H: Tumor-selective cytotoxicity of benzo[c] phenanthridine derivatives from Toddalia asiatica Lam. Cancer Chemother Pharmacol. 2010, 65: 719-726. 10.1007/s00280-009-1077-7.CrossRefPubMed Iwasaki H, Okabe T, Takara K, Toda T, Shimatani M, Oku H: Tumor-selective cytotoxicity of benzo[c] phenanthridine derivatives from Toddalia asiatica Lam. Cancer Chemother Pharmacol. 2010, 65: 719-726. 10.1007/s00280-009-1077-7.CrossRefPubMed
31.
Zurück zum Zitat Ingle WL, Salisbury JL: The role of the centrosomes in the development of malignant tumors. Current topics in Dev Biol. 1999, 49: 313-329.CrossRef Ingle WL, Salisbury JL: The role of the centrosomes in the development of malignant tumors. Current topics in Dev Biol. 1999, 49: 313-329.CrossRef
32.
Zurück zum Zitat Ganem NJ, Godunov SA, Pellman D: A mechanism linking extra centrosomes to chromosomal instability. Nature. 2009, 460: 278-282. 10.1038/nature08136.CrossRefPubMedPubMedCentral Ganem NJ, Godunov SA, Pellman D: A mechanism linking extra centrosomes to chromosomal instability. Nature. 2009, 460: 278-282. 10.1038/nature08136.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Fukasaw K: Oncogenes and tumour suppressors take in centrosomes. Nature Rev/Cancer. 2007, 7: 911-924. 10.1038/nrc2249. Fukasaw K: Oncogenes and tumour suppressors take in centrosomes. Nature Rev/Cancer. 2007, 7: 911-924. 10.1038/nrc2249.
34.
Zurück zum Zitat Fielding AB, Lim S, Montgomery K, Dobreva I, Dedhar S: critical role of integrin-linked kinase, ch-TOG and TACC3 in centrosome clustering in cancer cells. Oncogene. 2011, 30: 521-534. 10.1038/onc.2010.431.CrossRefPubMed Fielding AB, Lim S, Montgomery K, Dobreva I, Dedhar S: critical role of integrin-linked kinase, ch-TOG and TACC3 in centrosome clustering in cancer cells. Oncogene. 2011, 30: 521-534. 10.1038/onc.2010.431.CrossRefPubMed
35.
Zurück zum Zitat Galimberti F, Thompson SL, Liu X, Li H, Memoli V, et al: Targeting the Cyclin E-Cdk-2 Complex Represses Lung Cancer Growth by Triggering Anaphase Catastrophe. Clin Canc Res. 2010, 16: 109-120. 10.1158/1078-0432.CCR-09-2151.CrossRef Galimberti F, Thompson SL, Liu X, Li H, Memoli V, et al: Targeting the Cyclin E-Cdk-2 Complex Represses Lung Cancer Growth by Triggering Anaphase Catastrophe. Clin Canc Res. 2010, 16: 109-120. 10.1158/1078-0432.CCR-09-2151.CrossRef
36.
Zurück zum Zitat Jordan MA, Wilson L: Microtubules as a target for anticancer drugs. Nat Rev Cancer. 2004, 4: 253-265. 10.1038/nrc1317.CrossRefPubMed Jordan MA, Wilson L: Microtubules as a target for anticancer drugs. Nat Rev Cancer. 2004, 4: 253-265. 10.1038/nrc1317.CrossRefPubMed
37.
Zurück zum Zitat Chen JG, Horwitz SB: Differential mitotic responses to microtubule-stabilizing and de-stabilizing drugs. Cancer Res. 2002, 62: 1935-1938.PubMed Chen JG, Horwitz SB: Differential mitotic responses to microtubule-stabilizing and de-stabilizing drugs. Cancer Res. 2002, 62: 1935-1938.PubMed
38.
Zurück zum Zitat Rebacz B, Larsen TO, Clausen MH, Rønnest MH, Löffler H, Ho AD, Krämer A: Identification of Griseofulvin as an inhibitor of centrosomal clustering in a phenotype-based screen. Canc Res. 2007, 67: 6342-6350. 10.1158/0008-5472.CAN-07-0663.CrossRef Rebacz B, Larsen TO, Clausen MH, Rønnest MH, Löffler H, Ho AD, Krämer A: Identification of Griseofulvin as an inhibitor of centrosomal clustering in a phenotype-based screen. Canc Res. 2007, 67: 6342-6350. 10.1158/0008-5472.CAN-07-0663.CrossRef
39.
Zurück zum Zitat Jackson JR, Denis R, Patrick DR, Mohammed M, Dar MM, Huang PS: Targeted anti-mitotic therapies: can we improve on tubulin agents?. Nature Rev Cancer. 2007, 7: 107-117. 10.1038/nrc2049.CrossRef Jackson JR, Denis R, Patrick DR, Mohammed M, Dar MM, Huang PS: Targeted anti-mitotic therapies: can we improve on tubulin agents?. Nature Rev Cancer. 2007, 7: 107-117. 10.1038/nrc2049.CrossRef
40.
Zurück zum Zitat Kozielski F, DeBonis S, Skoufias DA: Screening for inhibitors of microtubule-associated motor proteins. Methods Mol Med. 2007, 137: 189-207. 10.1007/978-1-59745-442-1_14.CrossRefPubMed Kozielski F, DeBonis S, Skoufias DA: Screening for inhibitors of microtubule-associated motor proteins. Methods Mol Med. 2007, 137: 189-207. 10.1007/978-1-59745-442-1_14.CrossRefPubMed
41.
Zurück zum Zitat Manning JA, Kuma S: A potential role for NEDD1 and the centrosome in senescence of mouse embryonic fibroblasts. Cell Death Dis. 2010, 1: e35-10.1038/cddis.2010.12.CrossRefPubMedPubMedCentral Manning JA, Kuma S: A potential role for NEDD1 and the centrosome in senescence of mouse embryonic fibroblasts. Cell Death Dis. 2010, 1: e35-10.1038/cddis.2010.12.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Riffell JL, Janicke RU, Roberge M: Caspase-3-Dependent Mitotic Checkpoint Inactivation by the Small-Molecule Inducers of Mitotic Slippage SU6656 and Geraldol. Mol Cancer Ther. 2011, 10: 839-849. 10.1158/1535-7163.MCT-10-0909.CrossRefPubMed Riffell JL, Janicke RU, Roberge M: Caspase-3-Dependent Mitotic Checkpoint Inactivation by the Small-Molecule Inducers of Mitotic Slippage SU6656 and Geraldol. Mol Cancer Ther. 2011, 10: 839-849. 10.1158/1535-7163.MCT-10-0909.CrossRefPubMed
43.
Zurück zum Zitat Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, Teague J, et al: Mutations of the B-RAF gene in human cancer. Nature. 2002, 417: 949-954. 10.1038/nature00766.CrossRefPubMed Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, Teague J, et al: Mutations of the B-RAF gene in human cancer. Nature. 2002, 417: 949-954. 10.1038/nature00766.CrossRefPubMed
44.
Zurück zum Zitat Bild AH, Potti A, Nevins JR: Linking oncogenic pathways with therapeutic opportunities. Nat Rev Cancer. 2006, 6: 734-741.CrossRef Bild AH, Potti A, Nevins JR: Linking oncogenic pathways with therapeutic opportunities. Nat Rev Cancer. 2006, 6: 734-741.CrossRef
45.
Zurück zum Zitat O'Connor MJ, Martin NM, Smith GC: Targeted cancer therapies based on the inhibition of DNA strand break repair. Oncogene. 2007, 26: 7816-7824. 10.1038/sj.onc.1210879.CrossRefPubMed O'Connor MJ, Martin NM, Smith GC: Targeted cancer therapies based on the inhibition of DNA strand break repair. Oncogene. 2007, 26: 7816-7824. 10.1038/sj.onc.1210879.CrossRefPubMed
Metadaten
Titel
A phenanthrene derived PARP inhibitor is an extra-centrosomes de-clustering agent exclusively eradicating human cancer cells
verfasst von
Asher Castiel
Leonid Visochek
Leonid Mittelman
Françoise Dantzer
Shai Izraeli
Malka Cohen-Armon
Publikationsdatum
01.12.2011
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2011
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/1471-2407-11-412

Weitere Artikel der Ausgabe 1/2011

BMC Cancer 1/2011 Zur Ausgabe

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

CUP-Syndrom: Künstliche Intelligenz kann Primärtumor finden

30.04.2024 Künstliche Intelligenz Nachrichten

Krebserkrankungen unbekannten Ursprungs (CUP) sind eine diagnostische Herausforderung. KI-Systeme können Pathologen dabei unterstützen, zytologische Bilder zu interpretieren, um den Primärtumor zu lokalisieren.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.