Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Sci Rep ; 9(1): 14867, 2019 10 16.
Article in English | MEDLINE | ID: mdl-31619723

ABSTRACT

We previously showed that curcumin, a phytopolyphenol found in turmeric (Curcuma longa), targets a series of enzymes in the ROS metabolic pathway, induces irreversible growth arrest, and causes apoptosis. In this study, we tested Pentagamavunon-1 (PGV-1), a molecule related to curcumin, for its inhibitory activity on tumor cells in vitro and in vivo. PGV-1 exhibited 60 times lower GI50 compared to that of curcumin in K562 cells, and inhibited the proliferation of cell lines derived from leukemia, breast adenocarcinoma, cervical cancer, uterine cancer, and pancreatic cancer. The inhibition of growth by PGV-1 remained after its removal from the medium, which suggests that PGV-1 irreversibly prevents proliferation. PGV-1 specifically induced prometaphase arrest in the M phase of the cell cycle, and efficiently induced cell senescence and cell death by increasing intracellular ROS levels through inhibition of ROS-metabolic enzymes. In a xenograft mouse model, PGV-1 had marked anti-tumor activity with little side effects by oral administration, whereas curcumin rarely inhibited tumor formation by this administration. Therefore, PGV-1 is a potential therapeutic to induce tumor cell apoptosis with few side effects and low risk of relapse.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Curcumin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Prometaphase/drug effects , Administration, Oral , Alcohol Oxidoreductases/antagonists & inhibitors , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Animals , Antineoplastic Agents, Phytogenic/chemistry , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Death/drug effects , Cell Division/drug effects , Cell Division/genetics , Cell Movement/drug effects , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Curcumin/analogs & derivatives , Glutathione S-Transferase pi/antagonists & inhibitors , Glutathione S-Transferase pi/genetics , Glutathione S-Transferase pi/metabolism , Glutathione Transferase/antagonists & inhibitors , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , HEK293 Cells , HeLa Cells , Humans , K562 Cells , Lactoylglutathione Lyase/antagonists & inhibitors , Lactoylglutathione Lyase/genetics , Lactoylglutathione Lyase/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , MCF-7 Cells , Mice, Nude , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , Peroxiredoxins/antagonists & inhibitors , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , Prometaphase/genetics , Reactive Oxygen Species/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
2.
Cancer Lett ; 445: 1-10, 2019 03 31.
Article in English | MEDLINE | ID: mdl-30583077

ABSTRACT

We previously demonstrated that some N-biphenylanilides caused cell-cycle arrest at G2/M transition in breast cancer cells. Among them we choose three derivatives, namely PTA34, PTA73 and RS35 for experimentation in solid tumor cell lines, classical Hodgkin Lymphoma (cHL) cell lines and bona fide normal cell lines. Almost all tumor cells were sensitive to compounds in the nanomolar range whereas, they were not cytotoxic to normal ones. Interestingly the compounds caused a strong G2/M phase arrest in cHL cell lines, thus, here we investigated whether they affected the integrity of microtubules in such cells. We found that they induced a long prometaphase arrest, followed by induction of apoptosis which involved mitochondria. PTA73 and RS35 induced the mitotic arrest through the fragmentation of microtubules which prevented the kinethocore-mitotic spindle interaction and the exit from mitosis. PTA34 is instead a tubulin-targeting agent because it inhibited the tubulin polymerization as vinblastine. As such, PTA34 maintained the Cyclin B1-CDK1 regulatory complex activated during the G2/M arrest while inducing the inactivation of Bcl-2 through phosphorylation in Ser70, the degradation of Mcl-1 and a strong activation of BIML and BIMS proapoptotic isoforms. In addition PTA34 exerted an antiangiogenic effect by suppressing microvascular formation.


Subject(s)
Antimitotic Agents/chemical synthesis , Biphenyl Compounds/chemical synthesis , Hodgkin Disease/metabolism , Nicotine/chemistry , Antimitotic Agents/chemistry , Antimitotic Agents/pharmacology , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacology , CDC2 Protein Kinase/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin B1/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Hodgkin Disease/drug therapy , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Structure , Prometaphase/drug effects
3.
Mol Biol Cell ; 29(22): 2632-2643, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30133342

ABSTRACT

When untransformed human cells spend >1.5 h in prometaphase under standard culture conditions, all daughters arrest in G1 despite normal division of their mothers. We investigate what happens during prolonged prometaphase that leads to daughter cell arrest in the absence of DNA damage. We find that progressive loss of anti-apoptotic MCL-1 activity and oxidative stress act in concert to partially activate the apoptosis pathway, resulting in the delayed death of some daughters and senescence for the rest. At physiological oxygen levels, longer prometaphase durations are needed for all daughters to arrest. Partial activation of apoptosis during prolonged prometaphase leads to persistent caspase activity, which activates the kinase cascade mediating the post-mitotic activation of p38. This in turn activates p53, and the consequent expression of p21stops the cell cycle. This mechanism can prevent cells suffering intractable mitotic defects, which modestly prolong mitosis but allow its completion without DNA damage, from producing future cell generations that are susceptible to the evolution of a transformed phenotype.


Subject(s)
Apoptosis , Prometaphase , Apoptosis/drug effects , Caspase Inhibitors/pharmacology , Caspases/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Enzyme Activation/drug effects , Humans , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Oxidative Stress/drug effects , Oxygen/pharmacology , Prometaphase/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
4.
J Cell Sci ; 131(7)2018 04 12.
Article in English | MEDLINE | ID: mdl-29555820

ABSTRACT

During the prometaphase stage of mitosis, the cell builds a bipolar spindle of microtubules that mechanically segregates sister chromatids between two daughter cells in anaphase. The spindle assembly checkpoint (SAC) is a quality control mechanism that monitors proper attachment of microtubules to chromosome kinetochores during prometaphase. Segregation occurs only when each chromosome is bi-oriented with each kinetochore pair attached to microtubules emanating from opposite spindle poles. Overexpression of the protein kinase Aurora A is a feature of various cancers and is thought to enable tumour cells to bypass the SAC, leading to aneuploidy. Here, we took advantage of a chemical and chemical-genetic approach to specifically inhibit Aurora A kinase activity in late prometaphase. We observed that a loss of Aurora A activity directly affects SAC function, that Aurora A is essential for maintaining the checkpoint protein Mad2 on unattached kinetochores and that inhibition of Aurora A leads to loss of the SAC, even in the presence of nocodazole or Taxol. This is a new finding that should affect the way Aurora A inhibitors are used in cancer treatments.This article has an associated First Person interview with the first authors of the paper.


Subject(s)
Aurora Kinase A/genetics , M Phase Cell Cycle Checkpoints/genetics , Mad2 Proteins/genetics , Prometaphase/genetics , Anaphase/genetics , Aurora Kinase A/antagonists & inhibitors , Azepines/pharmacology , Cell Line, Tumor , Chromatids/genetics , Chromosome Segregation/drug effects , Gene Expression Regulation, Enzymologic/genetics , Humans , Kinetochores/drug effects , Microtubules/drug effects , Mitosis/drug effects , Mitosis/genetics , Nocodazole/pharmacology , Paclitaxel/pharmacology , Prometaphase/drug effects , Pyrimidines/pharmacology , Spindle Apparatus/genetics
5.
Cancer Lett ; 403: 74-85, 2017 09 10.
Article in English | MEDLINE | ID: mdl-28602975

ABSTRACT

Neuroblastoma is a biologically and clinically heterogeneous pediatric malignancy that includes a high-risk subset for which new therapeutic agents are urgently required. As well as MYCN amplification, activating point mutations of ALK and NRAS are associated with high-risk and relapsing neuroblastoma. As both ALK and RAS signal through the MEK/ERK pathway, we sought to evaluate two previously reported inhibitors of ETS-related transcription factors, which are transcriptional mediators of the Ras-MEK/ERK pathway in other cancers. Here we show that YK-4-279 suppressed growth and triggered apoptosis in nine neuroblastoma cell lines, while BRD32048, another ETV1 inhibitor, was ineffective. These results suggest that YK-4-279 acts independently of ETS-related transcription factors. Further analysis reveals that YK-4-279 induces mitotic arrest in prometaphase, resulting in subsequent cell death. Mechanistically, we show that YK-4-279 inhibits the formation of kinetochore microtubules, with treated cells showing a broad range of abnormalities including multipolar, fragmented and unseparated spindles, together leading to disrupted progression through mitosis. Notably, YK-4-279 does not affect microtubule acetylation, unlike the conventional mitotic poisons paclitaxel and vincristine. Consistent with this, we demonstrate that YK-4-279 overcomes vincristine-induced resistance in two neuroblastoma cell-line models. Furthermore, combinations of YK-4-279 with vincristine, paclitaxel or the Aurora kinase A inhibitor MLN8237/Alisertib show strong synergy, particularly at low doses. Thus, YK-4-279 could potentially be used as a single-agent or in combination therapies for the treatment of high-risk and relapsing neuroblastoma, as well as other cancers.


Subject(s)
Antimitotic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Indoles/pharmacology , Mitosis/drug effects , Neuroblastoma/drug therapy , Apoptosis/drug effects , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/metabolism , Azepines/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Synergism , Humans , Inhibitory Concentration 50 , Kinetochores/drug effects , Kinetochores/pathology , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , Paclitaxel/pharmacology , Prometaphase/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , RNA Interference , Signal Transduction/drug effects , Spindle Apparatus/drug effects , Spindle Apparatus/pathology , Time Factors , Transfection , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Vincristine/pharmacology
6.
Bioorg Med Chem Lett ; 27(12): 2731-2735, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28457756

ABSTRACT

In the course of our continuing studies on the 2-(benzo[b]thiophene-3'-yl)-6,8,8-triethyldesmosdumotin B (TEDB-TB) series, we designed and synthesized nine amino-TEDB-TB derivatives to improve pharmaceutical properties, identify structure activity relationships, and discover novel antitubulin agents. Among all newly synthesized amino-TEDB-TBs, the 5'- and 6'-amino derivatives, 6 and 7, exhibited significant antiproliferative activity against five human tumor cell lines, including an MDR subline overexpressing P-gp. The IC50 values of 0.50-1.01µM were 3-6 times better than those of previously reported hydroxy-TEDB-TBs. Compounds 6 and 7 inhibited tubulin polymerization, induced both depolymerization of interphase microtubules and multiple spindle formations, and caused cell arrest at prometaphase. Among all compounds, compound 7 scored best pharmaceutically with LogP 2.11 and biologically with greater antiproliferative activity and induction of cell cycle arrest at prometaphase.


Subject(s)
Antineoplastic Agents/pharmacology , Chromones/pharmacology , Flavonoids/pharmacology , Thiophenes/pharmacology , Tubulin/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chromones/chemical synthesis , Chromones/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Flavonoids/chemical synthesis , Flavonoids/chemistry , Humans , Molecular Structure , Polymerization/drug effects , Prometaphase/drug effects , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/chemistry , Tubulin Modulators
7.
Int J Oncol ; 49(4): 1305-14, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27498704

ABSTRACT

Resveratrol is a natural dietary product that has demonstrated multifaceted anticancer activity. Several analogues of resveratrol have been synthesized in an effort to enhance the pharmacological potency and improve the pharmacokinetic properties of the compound. 3,4,5,4'­trans­tetramethoxystilbene (3,4,5,4'­TMS) is a methoxylated analogue of resveratrol that has demonstrated anti-proliferative activity in vitro (in cancer cell lines) and in vivo (in xenograft models). In the present study, the anticancer effects of 3,4,5,4'­TMS in A375 human melanoma cells were examined. 3,4,5,4'­TMS markedly inhibited the proliferation of A375 cells (IC50=0.7 µM), via a mechanism involving mitotic arrest at the prometaphase stage of cell division. This effect was accompanied by the upregulation of the expression of the mitogen activated protein kinases, JNK and p38, and the concomitant activation of p38, that was verified by the nuclear translocation of the phoshorylated form of the protein. The pharmacological inhibition of p38 by SB203580 (4 µM) attenuated the effects of 3,4,5,4'­TMS, as demonstrated by decreased cell cycle progression at the mitotic phase. Furthermore, 3,4,5,4'­TMS increased the total levels of Aurora A, while it inhibited the localization of the protein to the spindle poles. Finally, 3,4,5,4'­TMS exhibited anti-metastatic activity, inhibiting A375 cell migration and the attachment of the cells to a collagen type IV-coated surface. Collectively, the data suggest that 3,4,5,4'­TMS is an effective chemotherapeutic drug for the treatment of human melanoma and that it exerts its effects through multiple anticancer modes of action.


Subject(s)
Antineoplastic Agents/pharmacology , MAP Kinase Signaling System/drug effects , Melanoma/metabolism , Stilbenes/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Melanoma/drug therapy , Prometaphase/drug effects
8.
Biochem Pharmacol ; 94(4): 257-69, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25732194

ABSTRACT

The present study sought to determine the correlation between 2-methoxyestradiol (2-MeO-E2)-induced cell cycle arrest and 2-MeO-E2-induced apoptosis. Exposure of Jurkat T cell clone (JT/Neo) to 2-MeO-E2 (0.5-1.0 µM) caused G2/M arrest, Bak activation, Δψm loss, caspase-9 and -3 activation, PARP cleavage, intracellular ROS accumulation, and apoptotic DNA fragmentation, whereas none of these events except for G2/M arrest were induced in Jurkat T cells overexpressing Bcl-2 (JT/Bcl-2). Under these conditions, Cdk1 phosphorylation at Thr-161 and dephosphorylation at Tyr-15, up-regulation of cyclin B1 expression, histone H1 phosphorylation, Cdc25C phosphorylation at Thr-48, Bcl-2 phosphorylation at Thr-56 and Ser-70, Mcl-1 phosphorylation at Ser-159/Thr-163, and Bim phosphorylation were detected irrespective of Bcl-2 overexpression. Concomitant treatment of JT/Neo cells with 2-MeO-E2 and the G1/S blocking agent aphidicolin resulted in G1/S arrest and abrogation of all apoptotic events, including Cdk1 activation, phosphorylation of Bcl-2, Mcl-1 and Bim, and ROS accumulation. The 2-MeO-E2-induced phosphorylation of Bcl-2 family proteins and mitochondrial apoptotic events were suppressed by a Cdk1 inhibitor, but not by an Aurora A kinase (AURKA), Aurora B kinase (AURKB), JNK, or p38 MAPK inhibitor. Immunofluorescence microscopic analysis revealed that 2-MeO-E2-induced mitotic arrest was caused by mitotic spindle network impairment and prometaphase arrest. Whereas 10-20 µM 2-MeO-E2 reduced the proportion of intracellular polymeric tubulin to monomeric tubulin, 0.5-5.0 µM 2-MeO-E2 increased it. These results demonstrate that the apoptogenic effect of 2-MeO-E2 (0.5-1.0 µM) was attributable to mitotic spindle defect-mediated prometaphase arrest, Cdk1 activation, phosphorylation of Bcl-2, Mcl-1, and Bim, and activation of Bak and mitochondria-dependent caspase cascade.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Estradiol/analogs & derivatives , Mitochondria/drug effects , Prometaphase/drug effects , Spindle Apparatus/drug effects , 2-Methoxyestradiol , Caspase 3/metabolism , Enzyme Activation , Estradiol/pharmacology , G1 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Jurkat Cells , Microtubules/drug effects , Microtubules/metabolism , Mitochondria/physiology , Mitosis , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Spindle Apparatus/physiology
9.
Biochem Biophys Res Commun ; 452(3): 845-51, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25218503

ABSTRACT

Exposure of human Jurkat T cells to JNK inhibitor IX (JNKi), targeting JNK2 and JNK3, caused apoptotic DNA fragmentation along with G2/M arrest, phosphorylation of Bcl-2, Mcl-1, and Bim, Δψm loss, and activation of Bak and caspase cascade. These JNKi-induced apoptotic events were abrogated by Bcl-2 overexpression, whereas G2/M arrest, cyclin B1 up-regulation, Cdk1 activation, and phosphorylation of Bcl-2 family proteins were sustained. In the concomitant presence of the G1/S blocking agent aphidicolin and JNKi, the cells underwent G1/S arrest and failed to induce all apoptotic events. The JNKi-induced phosphorylation of Bcl-2 family proteins and mitochondrial apoptotic events were suppressed by the Cdk1 inhibitor. Immunofluorescence microscopic analysis revealed that mitotic spindle defect and prometaphase arrest were the underlying factors for the G2/M arrest. These results demonstrate that JNKi-induced mitochondrial apoptosis was caused by microtubule damage-mediated prometaphase arrest, prolonged Cdk1 activation, and phosphorylation of Bcl-2 family proteins in Jurkat T cells.


Subject(s)
Mitochondria/drug effects , Mitogen-Activated Protein Kinase 10/antagonists & inhibitors , Mitogen-Activated Protein Kinase 9/antagonists & inhibitors , Prometaphase/drug effects , Protein Kinase Inhibitors/pharmacology , Aphidicolin/pharmacology , Apoptosis/drug effects , DNA Fragmentation/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation , Humans , Jurkat Cells , Membrane Potential, Mitochondrial/drug effects , Microtubules/drug effects , Mitochondria/metabolism , Mitogen-Activated Protein Kinase 10/genetics , Mitogen-Activated Protein Kinase 10/metabolism , Mitogen-Activated Protein Kinase 9/genetics , Mitogen-Activated Protein Kinase 9/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction
10.
Biochimie ; 104: 147-55, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24952350

ABSTRACT

Pterocarpans, a family of isoflavonoids found in the diverse Fabaceae, display potent cytotoxic activity over a panel of tumor cell lines, and among those tested, 2,3,9- trimethoxypterocarpan displays the most potent activity. This study evaluates the effects of 2,3,9-trimethoxypterocarpan and its related derivatives on cell cycle progression and microtubule function in select breast cancer cell lines (MCF7, T47d and HS578T). The pterocarpans, with the exception of 3,4-dihydroxy-9-methoxipterocarpan, induced increased frequencies of mitotic cells by inducing arrest in prometaphase. While microtubule organization in interphase cells was not modified during treatment, mitotic cells exhibited high frequencies of monastral spindles surrounded by condensed chromosomes. Immunofluorescence staining with an anti-γ-tubulin antibody showed double-dot labeling in the spindle polar region, suggesting that pterocarpan treatment blocked centrosome segregation. We found that this mitotic arrest was reversible when the cells were treated for up to 24 h followed by recovery in drug-free medium, but not after 48-h treatment followed by incubation in drug-free medium. In that case, treated cells typically underwent cell multinucleation and apoptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle Checkpoints/drug effects , Prometaphase/drug effects , Pterocarpans/pharmacology , Antineoplastic Agents/chemistry , Cell Death/drug effects , Cell Line, Tumor , Humans , Membrane Potential, Mitochondrial/drug effects , Prophase/drug effects , Pterocarpans/chemistry , Time Factors
11.
Cancer Biol Ther ; 15(7): 895-905, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24755487

ABSTRACT

Microtubules are essential cytoskeletal components with a central role in mitosis and have been particularly useful as a cancer chemotherapy target. We synthesized a small molecule derivative of a symmetrical 1,3-phenyl bis-thiourea, (1,1'-[1,3-phenylene]bis[3-(3,5-dimethylphenyl)thiourea], named "41J"), and identified a potent effect of the compound on cancer cell survival. 41J is cytotoxic to multiple cancer cell lines at nanomolar concentrations. Cell death occurred by apoptosis and was preceded by mitotic arrest in prometaphase. Prometaphase arrest induced by 41J treatment was accompanied by dissociation of cyclin B1 levels from the apparent mitotic stage and by major spindle abnormalities. Polymerization of purified tubulin in vitro was directly inhibited by 41J, suggesting that the compound works by directly interfering with microtubule function. Compound 41J arrested the growth of glioblastoma multiforme xenografts in nude mice at doses that were well-tolerated, demonstrating a relatively specific antitumor effect. Importantly, 41J overcame drug resistance due to ß-tubulin mutation and P-glycoprotein overexpression. Compound 41J may serve as a useful new lead compound for anticancer therapy development.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Microtubules/metabolism , Thiourea/analogs & derivatives , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor/drug effects , Central Nervous System Neoplasms/drug therapy , Cricetulus , Cyclin B1/metabolism , Drug Resistance, Neoplasm , Glioblastoma/drug therapy , Heterografts , Humans , Mice, Nude , Polymerization , Prometaphase/drug effects , Spindle Apparatus/drug effects , Thiourea/chemical synthesis , Thiourea/pharmacology , Thiourea/therapeutic use , Tubulin/metabolism
12.
Biochem Pharmacol ; 88(3): 291-302, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24486569

ABSTRACT

PM060184 belongs to a new family of tubulin-binding agents originally isolated from the marine sponge Lithoplocamia lithistoides. This compound is currently produced by total synthesis and is under evaluation in clinical studies in patients with advanced cancer diseases. It was recently published that PM060184 presents the highest known affinities among tubulin-binding agents, and that it targets tubulin dimers at a new binding site. Here, we show that PM060184 has a potent antitumor activity in a panel of different tumor xenograft models. Moreover, PM060184 is able to overcome P-gp mediated resistance in vivo, an effect that could be related to its high binding affinity for tubulin. To gain insight into the mechanism responsible of the observed antitumor activity, we have characterized its molecular and cellular effects. We have observed that PM060184 is an inhibitor of tubulin polymerization that reduces microtubule dynamicity in cells by 59%. Interestingly, PM060184 suppresses microtubule shortening and growing at a similar extent. This action affects cells in interphase and mitosis. In the first case, the compound induces a disorganization and fragmentation of the microtubule network and the inhibition of cell migration. In the second case, it induces the appearance of multipolar mitosis and lagging chromosomes at the metaphase plate. These effects correlate with prometaphase arrest and induction of caspase-dependent apoptosis or appearance of cells in a multinucleated interphase-like state unrelated to classical apoptosis pathways. Taken together, these results indicate that PM060184 represents a new tubulin binding agent with promising potential as an anticancer agent.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents/pharmacology , Polyketides/pharmacology , Pyrones/pharmacology , Tubulin Modulators/pharmacology , Animals , Apoptosis , Cell Line, Tumor , Cell Movement/drug effects , Centrosome/drug effects , Centrosome/ultrastructure , Female , Interphase , Mice, Nude , Microtubules/drug effects , Microtubules/ultrastructure , Prometaphase/drug effects , Spindle Apparatus/drug effects , Spindle Apparatus/ultrastructure , Xenograft Model Antitumor Assays
13.
Apoptosis ; 19(1): 224-40, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24166139

ABSTRACT

Treatment of Jurkat T cells with the microtubule-depolymerizing agent nocodazole (NOC) caused prometaphase arrest and apoptosis. NOC-induced mitochondrial apoptotic events including Bak activation, Δψm loss, cytochrome c release, and caspase cascade activation were blocked by Bcl-2 overexpression. However, mitotic arrest, Cdc25C activation, upregulation of cyclin B1 levels, Cdk1 activation, Bcl-2 phosphorylation at Thr-56 and Ser-70, and Bim phosphorylation were retained. The treatment of Jurkat T cells concomitantly with NOC and the G1/S-blocking agent hydroxyurea resulted in G1/S arrest and complete abrogation of all apoptotic events. The association of Bcl-2 with Bim or Bak declined after the prometaphase arrest-dependent phosphorylation of Bcl-2 and Bim, whereas the association of Bcl-2 with Bax remained relatively constant. Although Bax was redistributed from the cytosol to the mitochondria, resulting in an increase in the mitochondrial level of Bax following NOC treatment, the subcellular localization of Bcl-2, Bim, Bak and apoptosis-inducing factor was confined to the mitochondrial fraction irrespective of NOC treatment. Experiments using selective caspase inhibitors showed that mitochondria-dependent activation of caspase-9 and -3 was crucial for NOC-induced apoptosis. NOC-induced phosphorylation of Bcl-2 and Bim, Δψm loss, and mitochondria-dependent apoptotic events were significantly suppressed by a Cdk1 inhibitor roscovitine, but not by the JNK inhibitor SP600125 or the p38 MAPK inhibitor SB203580. These results show that the prometaphase arrest-dependent phosphorylation of Bcl-2 and Bim, which was mediated by Cdk1, could reduce the association of Bcl-2 with Bak or Bim to allow Bak activation and mitochondrial apoptotic events in Jurkat T cells exposed to NOC.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Jurkat Cells/drug effects , Leukemia, T-Cell/enzymology , Membrane Proteins/metabolism , Nocodazole/pharmacology , Prometaphase/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Caspase 3/genetics , Caspase 3/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cell Line, Tumor , Humans , Jurkat Cells/cytology , Jurkat Cells/metabolism , Leukemia, T-Cell/drug therapy , Leukemia, T-Cell/metabolism , Leukemia, T-Cell/physiopathology , M Phase Cell Cycle Checkpoints/drug effects , Membrane Proteins/genetics , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , bcl-2 Homologous Antagonist-Killer Protein/genetics
14.
J Cell Sci ; 127(Pt 2): 400-10, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24259668

ABSTRACT

Small GTP-binding proteins of the Rho family orchestrate the cytoskeleton remodelling events required for cell division. Guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs) promote cycling of Rho GTPases between the active GTP-bound and the inactive GDP-bound conformations. We report that ARHGAP19, a previously uncharacterised protein, is predominantly expressed in hematopoietic cells and has an essential role in the division of T lymphocytes. Overexpression of ARHGAP19 in lymphocytes delays cell elongation and cytokinesis. Conversely, silencing of ARHGAP19 or expression of a GAP-deficient mutant induces precocious mitotic cell elongation and cleavage furrow ingression, as well as excessive blebbing. In relation to these phenotypes, we show that ARHGAP19 acts as a GAP for RhoA, and controls recruitment of citron and myosin II to the plasma membrane of mitotic lymphocytes as well as Rock2-mediated phosphorylation of vimentin, which is crucial to maintain the stiffness and shape of lymphocytes. In addition to its effects on cell shape, silencing of ARHGAP19 in lymphocytes also impairs chromosome segregation.


Subject(s)
Chromosome Segregation , Cytokinesis , GTPase-Activating Proteins/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Shape/drug effects , Chromosome Segregation/drug effects , Cytokinesis/drug effects , Gene Expression Regulation, Leukemic/drug effects , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Leukemia/genetics , Leukemia/pathology , Mitosis/drug effects , Mitosis/genetics , Myosin Type II/metabolism , Nocodazole/pharmacology , Oligonucleotide Array Sequence Analysis , Phosphorylation/drug effects , Prometaphase/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein Transport/drug effects , Signal Transduction/drug effects , T-Lymphocytes/drug effects , Time Factors , Vimentin/metabolism , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
15.
Mol Carcinog ; 53(2): 125-37, 2014 Feb.
Article in English | MEDLINE | ID: mdl-22949227

ABSTRACT

Treatment of cancer cells with microtubule inhibitors causes mitotic arrest, which subsequently leads to cell death via activation of the intrinsic apoptotic pathway. Mitotically arrested cells typically display increased phosphorylation (i.e., inactivation) of two key anti-apoptotic proteins, Bcl-2 and Bcl-XL , but the mechanisms that regulate their phosphorylation as well as their role in apoptotic cell death following mitotic arrest are still poorly understood at present, which are the focus of this study. We recently showed that cyclin B1 and cell division cycle 2 (Cdc2) proteins are strongly up-regulated in human breast cancer cells following treatment with nocodazole (a prototypical microtubule inhibitor), and their up-regulation plays a critical role in the development of mitotic prometaphase arrest. In this study, we present evidence showing that the up-regulated cyclin B1/Cdc2 complex in nocodazole-treated human breast cancer cells is also responsible for the increased phosphorylation of Bcl-2 and Bcl-XL . However, only the increased phosphorylation of Bcl-XL , but not the phosphorylation of Bcl-2, contributes to subsequent activation of the intrinsic cell death pathway. In addition, evidence is presented to show that mitotic arrest deficient 2 (MAD2) is a key upstream mediator of the up-regulation of cyclin B1/Cdc2 as well as the subsequent increase in phosphorylationof Bcl-2 and Bcl-XL in nocodazole-treated cancer cells. Together, these results reveal that the up-regulated cyclin B1/Cdc2 complex not only mediates prometaphase arrest in nocodazole-treated cells, but also activates the subsequent intrinsic cell death pathway in these cells via increased phosphorylation of Bcl-XL .


Subject(s)
Apoptosis/genetics , Cell Cycle Checkpoints/genetics , Cyclin B1/genetics , Cyclin B/genetics , Mitosis/genetics , Phosphorylation/genetics , bcl-X Protein/genetics , Apoptosis/drug effects , CDC2 Protein Kinase , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cyclin-Dependent Kinases , Humans , MCF-7 Cells , Mad2 Proteins/genetics , Mitosis/drug effects , Nocodazole/pharmacology , Phosphorylation/drug effects , Prometaphase/drug effects , Prometaphase/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Up-Regulation/drug effects
16.
Biochim Biophys Acta ; 1833(10): 2220-32, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23707954

ABSTRACT

In Jurkat T cell clone (JT/Neo), G2/M arrest, apoptotic sub-G1 peak, mitochondrial membrane potential (Δψm) loss, and TUNEL-positive DNA fragmentation were induced following exposure to 17α-estradiol (17α-E2), whereas none of these events (except for G2/M arrest) were induced in Jurkat cells overexpressing Bcl-2 (JT/Bcl-2). Under these conditions, phosphorylation at Thr161 and dephosphorylation at Tyr15 of Cdk1, upregulation of cyclin B1 level, histone H1 phosphorylation, Cdc25C phosphorylation at Thr-48, Bcl-2 phosphorylation at Thr-56 and Ser-70, Mcl-1 phosphorylation, and Bim phosphorylation were detected in the presence of Bcl-2 overexpression. However, the 17α-E2-induced upregulation of Bak levels, activation of Bak, activation of caspase-3, and PARP degradation were abrogated by Bcl-2 overexpression. In the presence of the G1/S blocking agent hydroxyurea, 17α-E2 failed to induce G2/M arrest and all apoptotic events including Cdk1 activation and phosphorylation of Bcl-2, Mcl-1 and Bim. The 17α-E2-induced phosphorylation of Bcl-2 family proteins and mitochondrial apoptotic events were suppressed by a Cdk1 inhibitor but not by aurora A and aurora B kinase inhibitors. Immunofluorescence microscopic analysis showed that an aberrant bipolar microtubule array, incomplete chromosome congression at the metaphase plate, and prometaphase arrest, which was reversible, were the underlying factors for 17α-E2-induced mitotic arrest. The in vitro microtubule polymerization assay showed that 17α-E2 could directly inhibit microtubule formation. These results show that the apoptogenic activity of 17α-E2 was due to the impaired mitotic spindle assembly causing prometaphase arrest and prolonged Cdk1 activation, the phosphorylation of Bcl-2, Mcl-1 and Bim, and the activation of Bak and mitochondria-dependent caspase cascade.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Estradiol/pharmacology , Membrane Proteins/metabolism , Mitochondria/drug effects , Prometaphase/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , Bcl-2-Like Protein 11 , Blotting, Western , Caspases/metabolism , Estrogens/pharmacology , Flow Cytometry , Fluorescent Antibody Technique , Humans , In Situ Nick-End Labeling , Jurkat Cells , Microtubules/drug effects , Mitochondria/metabolism , Myeloid Cell Leukemia Sequence 1 Protein , Phosphorylation/drug effects
17.
Carcinogenesis ; 34(8): 1870-80, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23615397

ABSTRACT

Cardiac glycosides as inhibitors of the sodium/potassium adenosine triphosphatase (sodium pump) have been reported to block cancer growth by inducing G2/M phase arrest in many cancer cells. However, no detailed studies have been performed to distinguish between these two phases of cardiac glycoside-arrested cells. Furthermore, the underlying mechanisms involved in this cell cycle arrest process are still not known. Here, we report that bufalin and other cardiac glycosides potently induce mitotic arrest by the downregulation of polo-like kinase 1 (Plk1) expression. Live-cell imaging results demonstrate that bufalin-treated cells exhibit a marked delay in entering prophase at an early stage and are then arrested at prometaphase or induced entry into apoptosis. This phenotypic change is attributed to the downregulation of Plk1. We also show that bufalin and the knockdown of sodium pump reduce Plk1, at least in part, through downregulation of the nuclear transcription factors, hypoxia-inducible factor-1α (HIF-1α) and nuclear factor-kappa B (NF-κB). These findings suggest that cardiac glycosides induce mitotic arrest and apoptosis through HIF-1α- and NF-κB-mediated downregulation of Plk1 expression, demonstrating that HIF-1α and NF-κB are critical targets of cardiac glycosides in exerting their anticancer action.


Subject(s)
Cardiac Glycosides/pharmacology , Cell Cycle Proteins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Cycle Proteins/genetics , Cell Division/drug effects , Cell Division/genetics , Cell Line, Tumor , Down-Regulation/drug effects , G2 Phase/drug effects , G2 Phase/genetics , HCT116 Cells , HT29 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Microtubules/genetics , Microtubules/metabolism , NF-kappa B/genetics , Prometaphase/drug effects , Prometaphase/genetics , Prophase/drug effects , Prophase/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Polo-Like Kinase 1
18.
Biochim Biophys Acta ; 1823(8): 1306-15, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22580043

ABSTRACT

Earlier studies showed that 2-methoxyestradiol (2ME(2)), an endogenous nonpolar metabolite of estradiol-17ß, is a strong inducer of G(2)/M cell cycle arrest (based on analysis of cellular DNA content) in human cancer cell lines. The present study sought to investigate the molecular mechanism underlying 2ME(2)-induced cell cycle arrest. We found that 2ME(2) can selectively induce mitotic prometaphase arrest, but not G(2) phase arrest, in cultured MDA-MB-435s and MCF-7 human breast cancer cells. During the induction of prometaphase arrest, there is a time-dependent initial up-regulation of cyclin B1 and Cdc2 proteins, occurring around 12-24h. The strong initial up-regulation of cyclin B1 and Cdc2 matches in timing the 2ME(2)-induced prometaphase arrest. The 2ME(2)-induced prometaphase arrest is abrogated by selective knockdown of cyclin B1 and Cdc2, or by pre-treatment of cells with roscovitine, an inhibitor of cyclin-dependent kinases, or by co-treatment of cells with cycloheximide, a protein synthesis inhibitor that was found to suppress the early up-regulation of cyclin B1 and Cdc2. In addition, we provided evidence showing that MAD2 and JNK1 are important upstream mediators of 2ME(2)-induced up-regulation of cyclin B1 and Cdc2 as well as the subsequent induction of mitotic prometaphase arrest. In conclusion, treatment of human cancer cells with 2ME(2) causes up-regulation of cyclin B1 and Cdc2, which then mediate the induction of mitotic prometaphase arrest.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin B1/genetics , Cyclin B/genetics , Estradiol/analogs & derivatives , Prometaphase/drug effects , Up-Regulation/drug effects , 2-Methoxyestradiol , Breast Neoplasms , CDC2 Protein Kinase , Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Nucleus Shape/drug effects , Cyclin B/metabolism , Cyclin B1/metabolism , Cyclin-Dependent Kinases , Estradiol/pharmacology , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Knockdown Techniques , Humans , Mad2 Proteins , Mitogen-Activated Protein Kinase 8/metabolism , RNA Interference , Repressor Proteins/metabolism
19.
Mol Cell ; 44(5): 710-20, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22152475

ABSTRACT

The spindle assembly checkpoint (SAC) restricts mitotic exit to cells that have completed chromosome-microtubule attachment. Cdc20 is a bifunctional protein. In complex with SAC proteins Mad2, BubR1, and Bub3, Cdc20 forms the mitotic checkpoint complex (MCC), which binds the anaphase-promoting complex (APC/C) and inhibits its mitotic exit-promoting activity. When devoid of SAC proteins, Cdc20 serves as an APC/C coactivator and promotes mitotic exit. During mitotic arrest, Cdc20 is continuously degraded via ubiquitin-dependent proteolysis and resynthesized. It is believed that this cycle keeps the levels of Cdc20 below a threshold above which Cdc20 would promote mitotic exit. We report that p31(comet), a checkpoint antagonist, is necessary for mitotic destabilization of Cdc20. p31(comet) depletion stabilizes the MCC, super-inhibits the APC/C, and delays mitotic exit, indicating that Cdc20 proteolysis in prometaphase opposes the checkpoint. Our studies reveal a homeostatic network in which checkpoint-sustaining and -repressing forces oppose each other during mitotic arrest and suggest ways for enhancing the sensitivity of cancer cells to antitubulin chemotherapeutics.


Subject(s)
Homeostasis , Mitosis , Adaptor Proteins, Signal Transducing/metabolism , Anaphase-Promoting Complex-Cyclosome , Cdc20 Proteins , Cell Cycle Proteins/metabolism , HeLa Cells , Homeostasis/drug effects , Humans , M Phase Cell Cycle Checkpoints/drug effects , Mitosis/drug effects , Nocodazole/pharmacology , Nuclear Proteins/metabolism , Prometaphase/drug effects , Ubiquitin-Protein Ligase Complexes/metabolism
20.
PLoS One ; 6(8): e24312, 2011.
Article in English | MEDLINE | ID: mdl-21918689

ABSTRACT

BACKGROUND: During a normal cell cycle, the transition from G2 phase to mitotic phase is triggered by the activation of the cyclin B1-dependent Cdc2 kinase. Here we report our finding that treatment of MCF-7 human breast cancer cells with nocodazole, a prototypic microtubule inhibitor, results in strong up-regulation of cyclin B1 and Cdc2 levels, and their increases are required for the development of mitotic prometaphase arrest and characteristic phenotypes. METHODOLOGY/PRINCIPAL FINDINGS: It was observed that there was a time-dependent early increase in cyclin B1 and Cdc2 protein levels (peaking between 12 and 24 h post treatment), and their levels started to decline after the initial increase. This early up-regulation of cyclin B1 and Cdc2 closely matched in timing the nocodazole-induced mitotic prometaphase arrest. Selective knockdown of cyclin B1or Cdc2 each abrogated nocodazole-induced accumulation of prometaphase cells. The nocodazole-induced prometaphase arrest was also abrogated by pre-treatment of cells with roscovitine, an inhibitor of cyclin-dependent kinases, or with cycloheximide, a protein synthesis inhibitor that was found to suppress cyclin B1 and Cdc2 up-regulation. In addition, we found that MAD2 knockdown abrogated nocodazole-induced accumulation of cyclin B1 and Cdc2 proteins, which was accompanied by an attenuation of nocodazole-induced prometaphase arrest. CONCLUSIONS/SIGNIFICANCE: These observations demonstrate that the strong early up-regulation of cyclin B1 and Cdc2 contributes critically to the rapid and selective accumulation of prometaphase-arrested cells, a phenomenon associated with exposure to microtubule inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Cyclin B1/metabolism , Cyclin B/metabolism , Mitosis/drug effects , Nocodazole/pharmacology , Prometaphase/drug effects , Blotting, Western , Breast Neoplasms/pathology , CDC2 Protein Kinase , Cell Line, Tumor , Cyclin-Dependent Kinases , Female , Flow Cytometry , Humans , Microscopy, Fluorescence , RNA, Small Interfering
SELECTION OF CITATIONS
SEARCH DETAIL
...