Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
2.
J Nat Med ; 78(2): 370-381, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38265612

ABSTRACT

The Wnt/ß-catenin signaling pathway plays important roles in several cancer cells, including cell proliferation and development. We previously succeeded in synthesizing a small molecule compound inhibiting the Wnt/ß-catenin signaling pathway, named LPD-01 (1), and 1 inhibited the growth of human colorectal cancer (HT-29) cells. In this study, we revealed that 1 inhibits the growth of HT-29 cells stronger than that of another human colorectal cancer (SW480) cells. Therefore, we have attempted to identify the target proteins of 1 in HT-29 cells. Firstly, we investigated the effect on the expression levels of the Wnt/ß-catenin signaling pathway-related proteins. As a result, 1 inhibited the expression of target proteins of Wnt/ß-catenin signaling pathway (c-Myc and Survivin) and their genes, whereas the amount of transcriptional co-activator (ß-catenin) was not decreased, suggesting that 1 inhibited the Wnt/ß-catenin signaling pathway without affecting ß-catenin. Next, we investigated the target proteins of 1 using magnetic FG beads. Chemical pull-down assay combined with mass spectrometry suggested that 1 directly binds to importin7. As expected, 1 inhibited the nuclear translocation of importin7 cargoes such as Smad2 and Smad3 in TGF-ß-stimulated HT-29 cells. In addition, the knockdown of importin7 by siRNA reduced the expression of target genes of Wnt/ß-catenin signaling pathway. These results suggest that importin7 is one of the target proteins of 1 for inhibition of the Wnt/ß-catenin signaling pathway.


Subject(s)
Colorectal Neoplasms , beta Catenin , Humans , beta Catenin/metabolism , Wnt Signaling Pathway , Cell Proliferation , Colorectal Neoplasms/drug therapy , Cell Line, Tumor
3.
Eur J Pharmacol ; 963: 176229, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38072041

ABSTRACT

Anti-mitotic drugs are clinically used as anti-cancer treatments. Polo-like kinase 1 (PLK1) is a promising target against cancer cell division due to its importance in the whole process of mitosis, and thus PLK1-targeting agents have been developed in the last few decades. Clinical trial studies show that several PLK1 inhibitors are generally well-tolerated. However, the response rates are limited; therefore, it is needed to improve the efficacy of those drugs. Here, we show that NVP-BHG712, an erythropoietin-producing human hepatocellular (Eph) signaling inhibitor, potentiates the growth-inhibitory effects of the PLK1 inhibitors BI2536 and BI6727 in cancer cells. This combination treatment strongly suppresses cancer spheroid formation. Moreover, the combination drastically arrests cells at mitosis by continuous activation of the spindle assembly checkpoint (SAC), thereby inducing apoptosis. SAC activation caused by the combination of NVP-BHG712 and BI2536 is due to the inhibition of centrosome maturation and separation. Although the inactivation level of the PLK1 kinase is comparable between BI2536 treatment alone and combination treatment, the combination treatment strongly inactivates MAPK signaling in mitosis. Since inhibition of MAPK signaling potentiates the efficacy of BI2536 treatment, inactivation of PLK1 kinase and MAPK signaling contributes to the strong inhibition of centrosome separation. These results suggest that Eph signal inhibition potentiates the effect of PLK1 inhibition, leading to strong mitotic arrest via SAC activation and the subsequent reduction of cancer cell survival. The combination of PLK1 inhibition and Eph signal inhibition will provide a new effective strategy for targeting cancer cell division.


Subject(s)
Erythropoietin , Neoplasms , Humans , Cell Cycle , Cell Cycle Proteins , Cell Line, Tumor , Erythropoietin/antagonists & inhibitors , Mitosis , Neoplasms/drug therapy , Protein Serine-Threonine Kinases , Polo-like Kinases/antagonists & inhibitors
4.
Cell Signal ; 109: 110764, 2023 09.
Article in English | MEDLINE | ID: mdl-37315749

ABSTRACT

c-Src tyrosine kinase plays roles in a wide range of signaling events and its increased activity is frequently observed in a variety of epithelial and non-epithelial cancers. v-Src, an oncogene first identified in the Rous sarcoma virus, is an oncogenic version of c-Src and has constitutively active tyrosine kinase activity. We previously showed that v-Src induces Aurora B delocalization, resulting in cytokinesis failure and binucleated cell formation. In the present study, we explored the mechanism underlying v-Src-induced Aurora B delocalization. Treatment with the Eg5 inhibitor (+)-S-trityl-L-cysteine (STLC) arrested cells in a prometaphase-like state with a monopolar spindle; upon further inhibition of cyclin-dependent kinase (CDK1) by RO-3306, cells underwent monopolar cytokinesis with bleb-like protrusions. Aurora B was localized to the protruding furrow region or the polarized plasma membrane 30 min after RO-3306 addition, whereas inducible v-Src expression caused Aurora B delocalization in cells undergoing monopolar cytokinesis. Delocalization was similarly observed in monopolar cytokinesis induced by inhibiting Mps1, instead of CDK1, in the STLC-arrested mitotic cells. Importantly, western blotting analysis and in vitro kinase assay revealed that v-Src decreased the levels of Aurora B autophosphorylation and its kinase activity. Furthermore, like v-Src, treatment with the Aurora B inhibitor ZM447439 also caused Aurora B delocalization at concentrations that partially inhibited Aurora B autophosphorylation. Given that phosphorylation of Aurora B by v-Src was not observed, these results suggest that v-Src causes Aurora B delocalization by indirectly suppressing Aurora B kinase activity.


Subject(s)
Cytokinesis , Quinolines , Humans , Aurora Kinase B/metabolism , Phosphorylation , Oncogenes , Mitosis , HeLa Cells
5.
Exp Cell Res ; 429(2): 113672, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37339729

ABSTRACT

Heat shock is a physiological and environmental stress that leads to the denaturation and inactivation of cellular proteins and is used in hyperthermia cancer therapy. Previously, we revealed that mild heat shock (42 °C) delays the mitotic progression by activating the spindle assembly checkpoint (SAC). However, it is unclear whether SAC activation is maintained at higher temperatures than 42 °C. Here, we demonstrated that a high temperature of 44 °C just before mitotic entry led to a prolonged mitotic delay in the early phase, which was shortened by the SAC inhibitor, AZ3146, indicating SAC activation. Interestingly, mitotic slippage was observed at 44 °C after a prolonged delay but not at 42 °C heat shock. Furthermore, the multinuclear cells were generated by mitotic slippage in 44 °C-treated cells. Immunofluorescence analysis revealed that heat shock at 44 °C reduces the kinetochore localization of MAD2, which is essential for mitotic checkpoint activation, in nocodazole-arrested mitotic cells. These results indicate that 44 °C heat shock causes SAC inactivation even after full activation of SAC and suggest that decreased localization of MAD2 at the kinetochore is involved in heat shock-induced mitotic slippage, resulting in multinucleation. Since mitotic slippage causes drug resistance and chromosomal instability, we propose that there may be a risk of cancer malignancy when the cells are exposed to high temperatures.


Subject(s)
Cell Cycle Proteins , M Phase Cell Cycle Checkpoints , Humans , Cell Cycle Proteins/genetics , Mad2 Proteins/genetics , Mad2 Proteins/metabolism , Temperature , Spindle Apparatus/metabolism , Heat-Shock Response , Mitosis
6.
Sci Rep ; 13(1): 2067, 2023 02 04.
Article in English | MEDLINE | ID: mdl-36739326

ABSTRACT

Mitotic progression requires the precise formation of spindle microtubules based on mature centrosomes. During the G2/M transition, centrosome maturation progresses, and associated microtubules bundle to form mitotic spindle fibers and capture the chromosomes for alignment at the cell equator. Mitotic kinases-induced phosphorylation signaling is necessary for these processes. Here, we identified SH2 domain-containing protein 4A (SH2D4A/PPP1R38) as a new mitotic regulator. SH2D4A knockdown delays mitotic progression. The time-lapse imaging analysis showed that SH2D4A specifically contributes to the alignment of chromosomes. The cold treatment assay and microtubule regrowth assay indicated that SH2D4A promotes microtubule nucleation to support kinetochore-microtubule attachment. This may be due to the centrosome maturation by SH2D4A via centrosomal recruitment of pericentriolar material (PCM) such as cep192, γ-tubulin, and PLK1. SH2D4A was found to be a negative regulator of PP1 phosphatase. Consistently, treatment with a PP1 inhibitor rescues SH2D4A-knockdown-induced phenotypes, including the microtubule nucleation and centrosomal recruitment of active PLK1. These results suggest that SH2D4A is involved in PCM recruitment to centrosomes and centrosome maturation through attenuation of PP1 phosphatases, accelerating the spindle formation and supporting mitotic progression.


Subject(s)
Centrosome , Mitosis , Humans , Centrosome/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Microtubules/metabolism , Spindle Apparatus/metabolism , Tubulin/metabolism
7.
Genes Environ ; 45(1): 5, 2023 Jan 19.
Article in English | MEDLINE | ID: mdl-36658662

ABSTRACT

BACKGROUND: Heat shock proteins (Hsps) are overexpressed in several tumors and contribute to cell proliferation, metastasis, and anticancer drug resistance. Therefore, Hsp inhibitors have enhanced cytotoxicity as chemotherapeutic agents and may be effective with a reduced dosage for tumor therapy to avoid side effects. RESULTS: Four new azaphilones, maximazaphilones I-IV (1-4), and three known compounds (5-7) have been isolated from the airborne-derived fungus Penicillium maximae. Inhibitory effects of isolated compounds against induction of Hsp105 were evaluated by the luciferase assay system using Hsp105 promoter. In this assay, 2-4, 6, and 7 significantly inhibited hsp105 promoter activity without cytotoxicity. In addition, all isolated compounds except for 5 significantly induced the death of Adriamycin (ADR)-treated HeLa cells. Interestingly, 1-4, 6, and 7 didn't show anti-proliferative and cell death-inducing activity without ADR. CONCLUSION: This study revealed the chemical structures of maximazaphilones I-IV (1-4) and the potency of azaphilones may be useful for cancer treatment and reducing the dose of anticancer agents. In addition, one of the mechanisms of cell death-inducing activity for 2-4, 6, and 7 was suggested to be inhibitory effects of Hsp105 expression.

8.
J Cell Biochem ; 122(12): 1958-1967, 2021 12.
Article in English | MEDLINE | ID: mdl-34617313

ABSTRACT

The mammalian HSP105/110 family consists of four members, including Hsp105 and Apg-1, which function as molecular chaperones. Recently, we reported that Hsp105 knockdown increases sensitivity to the DNA-damaging agent Adriamycin but decreases sensitivity to the microtubule-targeting agent paclitaxel. However, whether the other Hsp105/110 family proteins have the same functional property is unknown. Here, we show that Apg-1 has different roles from Hsp105 in cell proliferation, cell division, and drug sensitivity. We generated the Apg-1-knockdown HeLa S3 cells by lentiviral expression of Apg-1-targeting short hairpin RNA. Knockdown of Apg-1 but not Hsp105 decreased cell proliferation. Apg-1 knockdown increased cell death upon Adriamycin treatment without affecting paclitaxel sensitivity. The cell synchronization experiment suggests that Apg-1 functions in mitotic progression at a different mitotic subphase from Hsp105, which cause difference in paclitaxel sensitivity. Since Apg-1 is overexpressed in certain types of tumors, Apg-1 may become a potential therapeutic target for cancer treatment without causing resistance to the microtubule-targeting agents.


Subject(s)
Cell Division , Drug Resistance, Neoplasm , HSP110 Heat-Shock Proteins/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , HSP110 Heat-Shock Proteins/genetics , HeLa Cells , Humans , Neoplasm Proteins/genetics , Neoplasms/genetics
9.
Fitoterapia ; 154: 105023, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34428520

ABSTRACT

Two new pyrrole alkaloids methyl-E-mangolamide (1) and methyl-Z-mangolamide (2), four new megastigmane glycosides nandinamegastigmanes I-IV (3-6), and eight known compounds (7-14) were isolated from the methanol extract of the fruits of Nandina domestica. The structures of the new compounds were elucidated based on chemical and spectroscopic evidence. The absolute stereochemistry of nandinamegastigmane I (3) was established upon comparing the experimental and predicted electronic circular dichroism (ECD) data. Among the isolated compounds, 1 and 2 showed cell death-inducing activity on the Adriamycin-treated HeLa cells. In addition, one of the mechanisms for cell death-inducing activity of 1 and 2 was suggested as inhibition of P-glycoprotein.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , Alkaloids/pharmacology , Berberidaceae/chemistry , Cell Death/drug effects , Alkaloids/isolation & purification , Fruit/chemistry , Glycosides/isolation & purification , HeLa Cells , Humans , Japan , Molecular Structure , Phytochemicals/isolation & purification , Phytochemicals/pharmacology
10.
Int J Mol Sci ; 22(11)2021 May 27.
Article in English | MEDLINE | ID: mdl-34071893

ABSTRACT

Insulin-like growth factor 1 receptor (IGF1R), a receptor-type tyrosine kinase, transduces signals related to cell proliferation, survival, and differentiation. We recently reported that OSI-906, an IGF1R inhibitor, in combination with the Aurora B inhibitor ZM447439 suppresses cell proliferation. However, the mechanism underlying this suppressive effect is yet to be elucidated. In this study, we examined the effects of combination treatment with OSI-906 and ZM447439 on cell division, so as to understand how cell proliferation was suppressed. Morphological analysis showed that the combination treatment generated enlarged cells with aberrant nuclei, whereas neither OSI-906 nor ZM447439 treatment alone caused this morphological change. Flow cytometry analysis indicated that over-replicated cells were generated by the combination treatment, but not by the lone treatment with either inhibitors. Time-lapse imaging showed mitotic slippage following a severe delay in chromosome alignment and cytokinesis failure with furrow regression. Furthermore, in S-trityl-l-cysteine-treated cells, cyclin B1 was precociously degraded. These results suggest that the combination treatment caused severe defect in the chromosome alignment and spindle assembly checkpoint, which resulted in the generation of over-replicated cells. The generation of over-replicated cells with massive aneuploidy may be the cause of reduction of cell viability and cell death. This study provides new possibilities of cancer chemotherapy.


Subject(s)
Aurora Kinase B/antagonists & inhibitors , Cell Survival/drug effects , Cyclin B1/metabolism , Imidazoles/pharmacology , Mitosis/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrazines/pharmacology , Benzamides/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Proteolysis , Quinazolines/pharmacology , Receptor, IGF Type 1/metabolism
11.
Bioorg Med Chem Lett ; 45: 128161, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34062253

ABSTRACT

Linderapyrone, a Wnt signal inhibitor was isolated from the methanolic extract of the stems and twigs of Lindera umbellata together with epi-(-)-linderol A. Linderapyrone inhibited TCF/ß-catenin transcriptional activity that was evaluated using cell-based TOPFlash luciferase assay system. To evaluate the structure-activity relationship and mechanism, we synthesized linderapyrone and its derivatives from piperitone. As the results of further bioassay for synthesized compounds, we found both of pyrone and monoterpene moieties were necessary for inhibitory effect. cDNA microarray analysis in a linderapyrone derivative treated human colorectal cancer cells showed that this compound downregulates Wnt signaling pathway. Moreover, we successes to synthesize the derivative of linderapyrone that has stronger inhibitory effect than linderapyrone and ICG-001 (positive control).


Subject(s)
Lindera/chemistry , TCF Transcription Factors/antagonists & inhibitors , beta Catenin/antagonists & inhibitors , Dose-Response Relationship, Drug , Humans , Molecular Structure , Structure-Activity Relationship , TCF Transcription Factors/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism
12.
J Cell Mol Med ; 25(3): 1677-1687, 2021 02.
Article in English | MEDLINE | ID: mdl-33465289

ABSTRACT

v-Src oncogene causes cell transformation through its strong tyrosine kinase activity. We have revealed that v-Src-mediated cell transformation occurs at a low frequency and it is attributed to mitotic abnormalities-mediated chromosome instability. v-Src directly phosphorylates Tyr-15 of cyclin-dependent kinase 1 (CDK1), thereby causing mitotic slippage and reduction in Eg5 inhibitor cytotoxicity. However, it is not clear whether v-Src modifies cytotoxicities of the other anticancer drugs targeting cell division. In this study, we found that v-Src restores cancer cell viability reduced by various microtubule-targeting agents (MTAs), although v-Src does not alter cytotoxicity of DNA-damaging anticancer drugs. v-Src causes mitotic slippage of MTAs-treated cells, consequently generating proliferating tetraploid cells. We further demonstrate that v-Src also restores cell viability reduced by a polo-like kinase 1 (PLK1) inhibitor. Interestingly, treatment with Aurora kinase inhibitor strongly induces cell death when cells express v-Src. These results suggest that the v-Src modifies cytotoxicities of anticancer drugs targeting cell division. Highly activated Src-induced resistance to MTAs through mitotic slippage might have a risk to enhance the malignancy of cancer cells through the increase in chromosome instability upon chemotherapy using MTAs.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Division/drug effects , Cell Survival/drug effects , Oncogene Protein pp60(v-src)/metabolism , Biomarkers , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , Flow Cytometry , Humans , Immunophenotyping , Microtubules/metabolism , Mitosis/drug effects , Mitosis/genetics , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Tubulin Modulators/pharmacology , Polo-Like Kinase 1
13.
FASEB J ; 35(1): e21242, 2021 01.
Article in English | MEDLINE | ID: mdl-33368671

ABSTRACT

When cells with excess DNA, such as tetraploid cells, undergo cell division, it can contribute to cellular transformation via asymmetrical chromosome segregation-generated genetic diversity. Cell cycle progression of tetraploid cells is suppressed by large tumor suppressor 2 (LATS2) kinase-induced inhibitory phosphorylation of the transcriptional coactivator Yes-associated protein (YAP). We recently reported that the oncogene v-Src induces tetraploidy and promotes cell cycle progression of tetraploid cells by suppressing LATS2 activity. We explore here the mechanism by which v-Src suppresses LATS2 activity and the role of LATS2 in v-Src-expressing cells. LATS2 was directly phosphorylated by v-Src and the proto-oncogene c-Src, resulting in decreased LATS2 kinase activity. This kinase-deficient LATS2 accumulated in a YAP transcriptional activity-dependent manner, and knockdown of either LATS2 or the LATS2-binding partner moesin-ezrin-radixin-like protein (Merlin) accelerated v-Src-induced membrane bleb formation. Upon v-Src expression, the interaction of Merlin with LATS2 was increased possibly due to a decrease in Merlin phosphorylation at Ser518, the dephosphorylation of which is required for the open conformation of Merlin and interaction with LATS2. LATS2 was colocalized with Merlin at the plasma membrane in a manner that depends on the Merlin-binding region of LATS2. The bleb formation in v-Src-expressing and LATS2-knockdown cells was rescued by the reexpression of wild-type or kinase-dead LATS2 but not the LATS2 mutant lacking the Merlin-binding region. These results suggest that the kinase-deficient LATS2 plays a role with Merlin at the plasma membrane in the maintenance of cortical rigidity in v-Src-expressing cells, which may cause tumor suppression.


Subject(s)
Cell Membrane Structures/enzymology , Oncogene Protein pp60(v-src)/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Membrane Structures/genetics , HCT116 Cells , HT29 Cells , HeLa Cells , Humans , Mice , NIH 3T3 Cells , Neurofibromin 2/genetics , Neurofibromin 2/metabolism , Oncogene Protein pp60(v-src)/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Mas , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , YAP-Signaling Proteins
14.
Exp Cell Res ; 395(2): 112207, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32750331

ABSTRACT

Cell division is a tightly regulated, essential process for cell proliferation. Very recently, we reported that EphA2 is phosphorylated at Ser897, via the Cdk1/MEK/ERK/RSK pathway, during M phase and contributes to proper M-phase progression by maintaining cortical rigidity via the EphA2pSer897/ephexin4/RhoG pathway. Here, we show that EphA2 kinase activity is dispensable for M-phase progression. Although EphA2 knockdown delayed this progression, the delay was rescued by an EphA2 mutant expression with an Asp739 to Asn substitution, as well as by wild-type EphA2. Western blotting analysis confirmed that the Asp739Asn mutant lost its EphA2 kinase activity. Like wild-type EphA2, the Asp739Asn mutant was localized to the plasma membrane irrespective of cell cycle. While RhoG localization to the plasma membrane was decreased in EphA2 knockdown cells, it was rescued by re-expression of wild-type EphA2 but not via the mutant containing the Ser897 to Ala substitution. This confirmed our recent report that phosphorylation at Ser897 is responsible for RhoG localization to the plasma membrane. In agreement with the M-phase progression's rescue effect, the Asp739Asn mutant rescued RhoG localization in EphA2 knockdown cells. These results suggest that EphA2 regulates M-phase progression in a manner independent of its kinase activity.


Subject(s)
Cell Cycle/physiology , Cell Division/physiology , Cell Proliferation/physiology , Ephrin-A2/metabolism , CDC2 Protein Kinase/metabolism , Guanine Nucleotide Exchange Factors/metabolism , HeLa Cells , Humans , MAP Kinase Signaling System/physiology , Phosphoserine/metabolism , Receptor, EphA2 , Signal Transduction/physiology
15.
J Nat Med ; 74(4): 689-701, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32535872

ABSTRACT

From the methanolic extract of the aerial parts of Petasites japonicus, six new eremophilane-type sesquiterpenoids, petasitesterpenes I-VI were isolated together with eight known compounds including S-japonin and eremophilenolide. The chemical structures of the isolated new compounds were elucidated based on chemical/physicochemical evidence. For petasitesterpenes I and II, the absolute configurations were established by comparison of experimental and predicted electronic circular dichroism (ECD) data. Among the isolated compounds, petasitesterpenes I, II, VI, and S-japonin showed cytotoxic activity against both human astrocytoma U-251MG cancer cells (non-CSCs) and their cancer stem cells (CSCs) isolated by sphere formation. In addition, cytotoxic activities of these compounds against breast cancer MDA-MB-231 were evaluated, supporting that petasitesterpene II has more effective than other isolated compounds.


Subject(s)
Neoplastic Stem Cells/chemistry , Petasites/chemistry , Plants, Medicinal/chemistry , Sesquiterpenes/chemistry , Humans , Molecular Structure
16.
Cancers (Basel) ; 12(4)2020 Apr 24.
Article in English | MEDLINE | ID: mdl-32344689

ABSTRACT

Anaplastic lymphoma kinase (ALK), a receptor-type tyrosine kinase, is involved in the pathogenesis of several cancers. ALK has been targeted with small molecule inhibitors for the treatment of different cancers, but absolute success remains elusive. In the present study, the effects of ALK inhibitors on M phase progression were evaluated. Crizotinib, ceritinib, and TAE684 suppressed proliferation of neuroblastoma SH-SY5Y cells in a concentration-dependent manner. At approximate IC50 concentrations, these inhibitors caused misorientation of spindles, misalignment of chromosomes and reduction in autophosphorylation. Similarly, knockdown of ALK caused M phase delay, which was rescued by re-expression of ALK. Time-lapse imaging revealed that anaphase onset was delayed. The monopolar spindle 1 (MPS1) inhibitor, AZ3146, and MAD2 knockdown led to a release from inhibitor-induced M phase delay, suggesting that spindle assembly checkpoint may be activated in ALK-inhibited cells. H2228 human lung carcinoma cells that express EML4-ALK fusion showed M phase delay in the presence of TAE684 at about IC50 concentrations. These results suggest that ALK plays a role in M phase regulation and ALK inhibition may contribute to the suppression of cell proliferation in ALK-expressing cancer cells.

17.
Fitoterapia ; 142: 104524, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32092530

ABSTRACT

Five new cadinene-type sesquiterpenes, hibiscusterpenes I-V (1-5), and six known compounds (6-11) have been isolated from the methanol extract of the stems and the twigs of Hibiscus tiliaceus. The structures of the new compounds were elucidated based on chemical and spectroscopic evidence. The absolute stereochemistry of hibiscusterpene I (1) was determined using X-ray crystallography. For hibiscusterpene III (3), the absolute configuration was established upon comparison of the experimental and predicted electronic circular dichroism (ECD) data. Among the isolated compounds, hibiscone C (7) and syriacusin A (11) showed cytotoxic activity in HeLa cells. In addition, their cell death-inducing activity was observed using time-lapse cell imaging.


Subject(s)
Antineoplastic Agents, Phytogenic/isolation & purification , Hibiscus/chemistry , Terpenes/isolation & purification , Antineoplastic Agents, Phytogenic/chemistry , Drug Screening Assays, Antitumor , HeLa Cells , Humans , Terpenes/chemistry
18.
Int J Mol Sci ; 21(3)2020 Feb 05.
Article in English | MEDLINE | ID: mdl-32033461

ABSTRACT

The insulin-like growth factor 1 receptor (IGF1R) is a receptor-type tyrosine kinase that transduces signals related to cell proliferation, differentiation, and survival. IGF1R expression is often misregulated in tumor cells, but the relevance of this for cancer progression remains unclear. Here, we examined the impact of IGF1R inhibition on cell division. We found that siRNA-mediated knockdown of IGF1R from HeLa S3 cells leads to M-phase delays. Although IGF1R depletion causes partial exclusion of FoxM1 from the nucleus, quantitative real-time PCR revealed that the transcription of M-phase regulators is not affected by decreased levels of IGF1R. Moreover, a similar delay in M phase was observed following 2 h of incubation with the IGF1R inhibitors OSI-906 and NVP-ADW742. These results suggest that the M-phase delay observed in IGF1R-compromised cells is not caused by altered expression of mitotic regulators. Live-cell imaging revealed that both prolonged prometaphase and prolonged metaphase underlie the delay and this can be abrogated by the inhibition of Mps1 with AZ3146, suggesting activation of the Spindle Assembly Checkpoint when IGF1R is inhibited. Furthermore, incubation with the Aurora B inhibitor ZM447439 potentiated the IGF1R inhibitor-induced suppression of cell proliferation, opening up new possibilities for more effective cancer chemotherapy.


Subject(s)
Aurora Kinase B/genetics , Cell Division/genetics , Cell Proliferation/genetics , Receptor, IGF Type 1/genetics , Apoptosis/drug effects , Apoptosis/genetics , Benzamides/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Division/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Disease Progression , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , HeLa Cells , Humans , Imidazoles/pharmacology , Pyrazines/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Quinazolines/pharmacology , Transcription, Genetic/drug effects , Transcription, Genetic/genetics
19.
J Cell Biochem ; 120(10): 17951-17962, 2019 10.
Article in English | MEDLINE | ID: mdl-31173393

ABSTRACT

Heat shock protein 105 (Hsp105) is a molecular chaperone, and the isoforms Hsp105α and Hsp105ß exhibit distinct functions with different subcellular localizations. Hsp105ß localizes in the nucleus and induces the expression of the major heat shock protein Hsp70, whereas cytoplasmic Hsp105α is less effective in inducing Hsp70 expression. Hsp105 shuttles between the cytoplasm and the nucleus; the subcellular localization is governed by the relative activities of the nuclear localization signal (NLS) and nuclear export signal (NES). Here, we show that nuclear accumulation of Hsp105α but not Hsp105ß is involved in Adriamycin (ADR) sensitivity. Knockdown of Hsp105α induces cell death at low ADR concentration, at which ADR is less effective in inducing cell death in the presence of Hsp105α. Of note, Hsp105 is localized in the nucleus under these conditions, even though Hsp105ß is not expressed, indicating that Hsp105α accumulates in the nucleus in response to ADR treatment. The exogenously expressed Hsp105α but not its NLS mutant localizes in the nucleus of ADR-treated cells. In addition, the expression level of the nuclear export protein chromosomal maintenance 1 (CRM1) was decreased by ADR treatment of cells, and CRM1 knockdown caused nuclear accumulation of Hsp105α both in the presence and absence of ADR. These results indicating that Hsp105α accumulates in the nucleus in a manner dependent on the NLS activity via the suppression of nuclear export. Our findings suggest a role of nuclear Hsp105α in the sensitivity against DNA-damaging agents in tumor cells.


Subject(s)
Cell Nucleus/metabolism , Doxorubicin/pharmacology , HSP110 Heat-Shock Proteins/metabolism , Nuclear Localization Signals/metabolism , Animals , COS Cells , Cell Death/drug effects , Cell Nucleus/drug effects , Cell Proliferation/drug effects , Chlorocebus aethiops , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Karyopherins/metabolism , Protein Transport/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Exportin 1 Protein
20.
Exp Cell Res ; 377(1-2): 67-74, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30776355

ABSTRACT

Thermotolerance is a phenomenon in which cells become resistant to stress by prior exposure to heat shock, and its development is associated with the induction of heat shock proteins (Hsps), including Hsp70. We previously showed that the expression of Hsp70 is regulated by the cytokine signaling transcription factor Stat3, but the role of Stat3 in thermotolerance is not known. In this study, we examined the possible involvement of Stat3 in the acquisition of thermotolerance. We found that severe heat shock-induced morphological changes and decreases in cell viability, which were suppressed by exposure to non-lethal mild heat shock prior to severe heat shock. This thermotolerance development was accompanied by Stat3 phosphorylation and the induction of Hsps such as Hsp105, Hsp70, and Hsp27. Stat3 phosphorylation and Hsp induction were inhibited by AG490, an inhibitor of JAK tyrosine kinase. Consistent with this, we found that mild heat shock-induced thermotolerance was partially suppressed by AG490 or knockdown of Hsp105. We also found that the Stat3 inhibitor Stattic suppresses the acquisition of thermotolerance by inhibiting the mild heat shock-induced Stat3 phosphorylation and Hsp105 expression. These results suggest that the mild heat shock-dependent stimulation of the JAK-Stat signaling pathway contributes to the development of thermotolerance via the induction of Hsps including Hsp105. This signaling pathway may be a useful target for hyperthermia cancer therapy.


Subject(s)
Heat-Shock Proteins/metabolism , Heat-Shock Response , Hot Temperature , Hyperthermia, Induced/methods , STAT3 Transcription Factor/metabolism , Thermotolerance , HeLa Cells , Humans , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL
...