Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 42
1.
ACS Appl Mater Interfaces ; 14(1): 20-31, 2022 Jan 12.
Article En | MEDLINE | ID: mdl-34914354

Tissue microarchitecture imposes physical constraints to the migration of individual cells. Especially in cancer metastasis, three-dimensional structural barriers within the extracellular matrix are known to affect the migratory behavior of cells, regulating the pathological state of the cells. Here, we employed a culture platform with micropillar arrays of 2 µm diameter and 16 µm pitch (2.16 micropillar) as a mechanical stimulant. Using this platform, we investigated how a long-term culture of A549 human lung carcinoma cells on the (2.16) micropillar-embossed dishes would influence the pathological state of the cell. A549 cells grown on the (2.16) micropillar array with 10 µm height exhibited a significantly elongated morphology and enhanced migration even after the detachment and reattachment, as evidenced in the conventional wound-healing assay, single-cell tracking analysis, and in vivo tumor colonization assays. Moreover, the pillar-induced morphological deformation in nuclei was accompanied by cell-cycle arrest in the S phase, leading to suppressed proliferation. While these marked traits of morphology-migration-proliferation support more aggressive characteristics of metastatic cancer cells, typical indices of epithelial-mesenchymal transition were not found, but instead, remarkable traces of amoeboidal transition were confirmed. Our study also emphasizes the importance of mechanical stimuli from the microenvironment during pathogenesis and how gained traits can be passed onto subsequent generations, ultimately affecting their pathophysiological behavior. Furthermore, this study highlights the potential use of pillar-based mechanical stimuli as an in vitro cell culture strategy to induce more aggressive tumorigenic cancer cell models.


Cell Culture Techniques/methods , Lung Neoplasms/metabolism , A549 Cells , Animals , Cell Culture Techniques/instrumentation , Cell Movement/physiology , Cell Proliferation/physiology , Fatty Acids/metabolism , Female , Humans , Mechanical Phenomena , Metabolomics , Mice, Inbred BALB C , Mice, Nude , S Phase Cell Cycle Checkpoints/physiology
2.
Cell Cycle ; 18(21): 2876-2892, 2019 Nov.
Article En | MEDLINE | ID: mdl-31522595

Glioblastoma is the most aggressive brain tumor. Although miR-141 has been demonstrated to primarily function as a tumor suppressor in numerous malignancies, including glioblastoma, the mechanisms involved remain poorly understood. Here, it is shown that miR-141 is downregulated in glioblastoma cell lines and tissues and may exert its biological function via directly targeting myelin transcription factor 1-like (MYT1L). Using two glioblastoma cell lines that differ from each other by the functionality of DNA-dependent protein kinase (DNAPK), a functional involvement of DNAPK in the miR-141 tumor suppression network was observed. In M059K cells with a normal function of DNAPK, the enforced expression of miR-141 attenuated MYT1L expression and suppressed cell proliferation. Conversely, the inhibition of miR-141 expression promoted cell proliferation; however, in M059J cells with a loss-of-function DNAPK, miR-141 constitutively inhibited cell proliferation upon ectopic overexpression or inhibition. An overexpression of miR-141 suppressed M059J cell migration, while it had no effect on M059K. Furthermore, the ectopic expression of miR-141 induced an S-phase arrest in both cell lines, whereas the inhibition of miR-141 caused a G1 arrest in M059J and accelerated the S phase in M059K. An overexpression and suppression of miR-141 resulted in an aberrant expression of cell-cycle proteins, including p21. Moreover, MYT1L may be a transcription factor of p21 in p53-mutant cells, whereas DNAPK may function as a repressor of MYT1L. The findings revealed the crucial role of DNAPK in miR-141-mediated suppression of gliomagenesis and demonstrated that it may be a target molecule in miR-141-associated therapeutic interventions for glioblastoma.


Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA-Activated Protein Kinase/metabolism , Glioblastoma/pathology , MicroRNAs/genetics , Nerve Tissue Proteins/metabolism , Transcription Factors/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA-Binding Proteins/metabolism , Genes, Tumor Suppressor/physiology , Glioblastoma/genetics , Glioblastoma/therapy , Humans , S Phase Cell Cycle Checkpoints/physiology
3.
Mol Biol Cell ; 30(22): 2771-2789, 2019 10 15.
Article En | MEDLINE | ID: mdl-31509480

Budding yeast treated with hydroxyurea (HU) activate the S phase checkpoint kinase Rad53, which prevents DNA replication forks from undergoing aberrant structural transitions and nuclease processing. Rad53 is also required to prevent premature extension of the mitotic spindle that assembles during a HU-extended S phase. Here we present evidence that checkpoint restraint of spindle extension is directly coupled to Rad53 control of replication fork stability. In budding yeast, centromeres are flanked by replication origins that fire in early S phase. Mutations affecting the Zn2+-finger of Dbf4, an origin activator, preferentially reduce centromere-proximal origin firing in HU, corresponding with suppression of rad53 spindle extension. Inactivating Exo1 nuclease or displacing centromeres from origins provides a similar suppression. Conversely, short-circuiting Rad53 targeting of Dbf4, Sld3, and Dun1, substrates contributing to fork stability, induces spindle extension. These results reveal spindle extension in HU-treated rad53 mutants is a consequence of replication fork catastrophes at centromeres. When such catastrophes occur, centromeres become susceptible to nucleases, disrupting kinetochore function and spindle force balancing mechanisms. At the same time, our data indicate centromere duplication is not required to stabilize S phase spindle structure, leading us to propose a model for how monopolar kinetochore-spindle attachments may contribute to spindle force balance in HU.


Caenorhabditis elegans Proteins/metabolism , DNA Replication/physiology , Protein Serine-Threonine Kinases/metabolism , Spindle Apparatus/metabolism , Cell Cycle Checkpoints , Cell Cycle Proteins/metabolism , Centromere/genetics , Centromere/metabolism , Checkpoint Kinase 2/genetics , Chromosome Segregation/drug effects , Chromosome Structures/metabolism , DNA Damage/genetics , DNA Replication/genetics , DNA, Fungal/genetics , Kinetochores/metabolism , Replication Origin , S Phase/physiology , S Phase Cell Cycle Checkpoints/genetics , S Phase Cell Cycle Checkpoints/physiology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
4.
BMC Mol Cell Biol ; 20(1): 8, 2019 04 11.
Article En | MEDLINE | ID: mdl-31041891

BACKGROUND: Dlx5 and Dlx6 stimulate differentiation of diverse progenitors during embryonic development. Their actions as pro-differentiation transcription factors includes the up-regulation of differentiation markers but the extent to which differentiation may also be stimulated by regulation of the cell cycle has not been addressed. RESULTS: We document that expression of Dlx5 and Dlx6 antagonizes cell proliferation in a variety of cell types without inducing apoptosis or promoting cell cycle exit. Rather, a variety of evidence indicates that elevated Dlx5 and Dlx6 expression reduces the proportion of cells in S phase and affects the length of the cell cycle. CONCLUSIONS: Antagonism of S-phase entry by Dlx5 and Dlx6 proteins likely represents a lineage-independent function to effect Dlx-mediated differentiation in multiple progenitor cell types.


Cell Division/physiology , G1 Phase Cell Cycle Checkpoints/physiology , Homeodomain Proteins/genetics , S Phase Cell Cycle Checkpoints/physiology , Transcription Factors/genetics , Animals , Apoptosis/physiology , Cell Differentiation/physiology , Cell Proliferation/physiology , Chick Embryo , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Mice , Plasmids/genetics , Transfection
5.
Life Sci Alliance ; 2(2)2019 04.
Article En | MEDLINE | ID: mdl-30979824

Human CTC1-STN1-TEN1 (CST) is an RPA-like single-stranded DNA-binding protein that interacts with DNA polymerase α-primase (pol α) and functions in telomere replication. Previous studies suggest that CST also promotes replication restart after fork stalling. However, the precise role of CST in genome-wide replication remains unclear. In this study, we sought to understand whether CST alters origin licensing and activation. Replication origins are licensed by loading of the minichromosome maintenance 2-7 (MCM) complex in G1 followed by replisome assembly and origin firing in S-phase. We find that CST directly interacts with the MCM complex and disrupts binding of CDT1 to MCM, leading to decreased origin licensing. We also show that CST enhances replisome assembly by promoting AND-1/pol α chromatin association. Moreover, these interactions are not dependent on exogenous replication stress, suggesting that CST acts as a specialized replication factor during normal replication. Overall, our findings implicate CST as a novel regulator of origin licensing and replisome assembly/fork progression through interactions with MCM, AND-1, and pol α.


Chromatin/metabolism , DNA Replication/physiology , DNA-Binding Proteins/metabolism , Telomere-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , DNA Polymerase I/metabolism , G1 Phase Cell Cycle Checkpoints/physiology , Gene Knockdown Techniques , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Minichromosome Maintenance Proteins/metabolism , RNA, Small Interfering/genetics , S Phase Cell Cycle Checkpoints/physiology , Telomere/metabolism , Telomere-Binding Proteins/genetics
6.
Life Sci Alliance ; 2(2)2019 04.
Article En | MEDLINE | ID: mdl-30988162

The precise coordination of growth and proliferation has a universal prevalence in cell homeostasis. As a prominent property, cell size is modulated by the coordination between these processes in bacterial, yeast, and mammalian cells, but the underlying molecular mechanisms are largely unknown. Here, we show that multifunctional chaperone systems play a concerted and limiting role in cell-cycle entry, specifically driving nuclear accumulation of the G1 Cdk-cyclin complex. Based on these findings, we establish and test a molecular competition model that recapitulates cell-cycle-entry dependence on growth rate. As key predictions at a single-cell level, we show that availability of the Ydj1 chaperone and nuclear accumulation of the G1 cyclin Cln3 are inversely dependent on growth rate and readily respond to changes in protein synthesis and stress conditions that alter protein folding requirements. Thus, chaperone workload would subordinate Start to the biosynthetic machinery and dynamically adjust proliferation to the growth potential of the cell.


Cell Enlargement , Cell Size , G1 Phase Cell Cycle Checkpoints/physiology , Heat-Shock Response/physiology , Molecular Chaperones/metabolism , Salt Stress/physiology , CDC28 Protein Kinase, S cerevisiae/metabolism , Cell Nucleolus/metabolism , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , HSP40 Heat-Shock Proteins/metabolism , Models, Molecular , S Phase Cell Cycle Checkpoints/physiology , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae Proteins/metabolism
7.
FEBS Open Bio ; 9(6): 1109-1118, 2019 06.
Article En | MEDLINE | ID: mdl-30972973

Bone morphogenetic protein 4 (BMP4) has been reported to regulate adipose development, but its role in preadipocyte proliferation has not been explored in vitro. Here, we investigated the effect of BMP4 on chicken preadipocyte proliferation using immortalized chicken preadipocytes (ICP1 cells) as a cell model. We report that BMP4 expression increases during preadipocyte proliferation. Overexpression and knockdown of BMP4 promotes and inhibits preadipocyte proliferation, respectively. In addition, overexpression of BMP4 decreased the number of preadipocytes at the G0/G1 phase of the cell cycle, and increased the proportion of cells at S phase. In contrast, knockdown of BMP4 increased the number of preadipocytes at the G0/G1 phase of the cell cycle, and decreased the proportion of cells at the S and G2 phases. Furthermore, overexpression of BMP4 promoted the expression of proliferating cell nuclear antigen (PCNA), Id2, cyclin E, and cyclin-dependent kinase 2 (CDK2), while knockdown of BMP4 inhibited the expression of Id2, cyclin E, and CDK2. Finally, neither BMP4 overexpression nor BMP4 knockdown affected cell apoptosis. Taken together, our results suggest that BMP4 may promote proliferation of ICP1 cells by driving cell cycle transition from G1 to S phase.


Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Cell Proliferation/physiology , Chickens , Fibroblasts/metabolism , G1 Phase Cell Cycle Checkpoints/physiology , S Phase Cell Cycle Checkpoints/physiology , Animals , Apoptosis , Cell Line , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/metabolism , Gene Knockdown Techniques , Inhibitor of Differentiation Protein 2/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Transfection , Up-Regulation
8.
Pharm Biol ; 56(1): 422-432, 2018 Dec.
Article En | MEDLINE | ID: mdl-30301390

CONTEXT: Dicranopteris linearis (Burm.f.) Underw. (Gleicheniaceae) has been scientifically proven to exert various pharmacological activities. Nevertheless, its anti-proliferative potential has not been extensively investigated. OBJECTIVE: To investigate the anti-proliferative potential of D. linearis leaves and determine possible mechanistic pathways. MATERIALS AND METHODS: MTT assay was used to determine the cytotoxic effects of D. linearis methanol (MEDL) and petroleum ether (PEEDL) extracts at concentrations of 100, 50, 25, 12.5, 6.25 and 3.125 µg/mL against a panel of cancer cell lines (breast [MCF-7 and MDA-MB-231], cervical [HeLa], colon [HT-29], hepatocellular [HepG2] and lung [A549]), as compared to negative (untreated) and positive [5-fluorouracil (5-FU)-treated] control groups. Mouse fibroblast cells (3T3) were used as normal cells. The mode of cell death was examined using morphological analysis via acridine orange (AO) and propidium iodide (PI) double staining. Cell cycle arrest was determined using flow cytometer, followed by annexin V-PI apoptosis detection kit. RESULTS: MEDL demonstrated the most significant growth inhibition against MDA-MB-231 cells (IC50 22.4 µg/mL). PEEDL showed no cytotoxic effect. Induction of apoptosis by MEDL was evidenced via morphological analysis and acridine orange propidium iodide staining. MEDL could induce S phase cell cycle arrest after 72 h of incubation. Early apoptosis induction in MDA-MB-231 cells was confirmed by annexin V-FITC and PI staining. Significant increase in apoptotic cells were detected after 24 h of treatment with 15.07% cells underwent apoptosis, and the amount escalated to 18.24% with prolonged 48 h incubation. CONCLUSIONS: MEDL has potential as a potent cytotoxic agent against MDA-MB-231 adenocarcinoma.


Apoptosis/drug effects , Breast Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Plant Extracts/pharmacology , S Phase Cell Cycle Checkpoints/drug effects , 3T3 Cells , A549 Cells , Animals , Apiaceae , Apoptosis/physiology , Breast Neoplasms/drug therapy , Cell Cycle Checkpoints/physiology , Cell Line, Tumor , Cell Proliferation/physiology , HeLa Cells , Hep G2 Cells , Humans , MCF-7 Cells , Mice , Plant Extracts/isolation & purification , S Phase Cell Cycle Checkpoints/physiology
9.
Mol Microbiol ; 110(2): 191-203, 2018 10.
Article En | MEDLINE | ID: mdl-30084240

Candida albicans is an opportunistic fungal pathogen. In immunocompromised individuals, it can cause bloodstream infections with high mortality rates. The ability to switch between yeast and hyphal morphologies is a critical virulence factor of C. albicans. In response to diverse environmental cues, several signaling pathways are activated resulting in filamentous growth. Interestingly, cell cycle arrest can also trigger filamentous growth although the pathways involved are not well-understood. Here, we demonstrate that the cAMP-PKA pathway is involved in the filamentous growth caused by G1 arrest due to the depletion of the G1 cyclin Cln3 and S phase arrest due to hydroxyurea treatment. The downstream mechanisms involved in filamentation are different between the two cell cycle arrest phenomena. Cln3-depleted cells require HGC1 and UME6 for filamentous growth, but hydroxyurea-induced filamentation does not. Also, the hyphal repressor Nrg1 is not involved in the suppression of Cln3-depletion and hydroxyurea-induced filamentous growth. The findings highlight the complexity of the signaling networks that control filamentous growth in which different mechanisms downstream of the cAMP-PKA pathway are activated based on the nature of the inducing signals.


Candida albicans/growth & development , Cyclins/metabolism , Fungal Proteins/metabolism , G1 Phase Cell Cycle Checkpoints/physiology , Hyphae/growth & development , S Phase Cell Cycle Checkpoints/physiology , Candida albicans/drug effects , Candida albicans/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclins/genetics , Fungal Proteins/genetics , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Deletion , Humans , Hydroxyurea/pharmacology , Hyphae/drug effects , Plasmids , Repressor Proteins/metabolism , S Phase Cell Cycle Checkpoints/drug effects
10.
Nat Commun ; 9(1): 2221, 2018 06 07.
Article En | MEDLINE | ID: mdl-29880867

Cell growth and survival depend on a delicate balance between energy production and synthesis of metabolites. Here, we provide evidence that an alternative mitochondrial complex II (CII) assembly, designated as CIIlow, serves as a checkpoint for metabolite biosynthesis under bioenergetic stress, with cells suppressing their energy utilization by modulating DNA synthesis and cell cycle progression. Depletion of CIIlow leads to an imbalance in energy utilization and metabolite synthesis, as evidenced by recovery of the de novo pyrimidine pathway and unlocking cell cycle arrest from the S-phase. In vitro experiments are further corroborated by analysis of paraganglioma tissues from patients with sporadic, SDHA and SDHB mutations. These findings suggest that CIIlow is a core complex inside mitochondria that provides homeostatic control of cellular metabolism depending on the availability of energy.


Electron Transport Complex II/metabolism , Energy Metabolism/physiology , Mitochondria/metabolism , Paraganglioma/pathology , Stress, Physiological , Animals , Biosynthetic Pathways/physiology , Cell Line, Tumor , Electron Transport Complex II/genetics , Gene Knockout Techniques , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , Paraganglioma/genetics , RNA, Small Interfering/metabolism , S Phase Cell Cycle Checkpoints/physiology , Succinate Dehydrogenase/genetics , Succinate Dehydrogenase/metabolism , Xenograft Model Antitumor Assays
11.
FASEB J ; 31(7): 2925-2936, 2017 07.
Article En | MEDLINE | ID: mdl-28360195

Polo-like kinase 1 (PLK1) is a serine/threonine kinase involved in several stages of the cell cycle, including the entry and exit from mitosis, and cytokinesis. Furthermore, it has an essential role in the regulation of DNA replication. Together with cyclin A, PLK1 also promotes CDH1 phosphorylation to trigger its ubiquitination and degradation, allowing cell cycle progression. The PLK1 levels in different type of tumors are very high compared to normal tissues, which is consistent with its role in promoting proliferation. Therefore, several PLK1 inhibitors have been developed and tested for the treatment of cancer. Here, we further analyzed PLK1 degradation and found that cytoplasmic PLK1 is ubiquitinated and subsequently degraded by the SCFßTrCP/proteasome. This procedure is triggered when heat shock protein (HSP) 90 is inhibited with geldanamycin, which results in misfolding of PLK1. We also identified CDK1 as the major kinase involved in this degradation. Our work shows for the first time that HSP90 inhibition arrests cell cycle progression at the G1/S transition. This novel mechanism inhibits CDH1 degradation through CDK1-dependent PLK1 destruction by the SCFßTrCP/proteasome. In these conditions, CDH1 substrates do not accumulate and cell cycle arrests, providing a novel pathway for regulation of the cell cycle at the G1-to-S boundary.-Giráldez, S., Galindo-Moreno, M., Limón-Mortés, M. C., Rivas, A. C., Herrero-Ruiz, J., Mora-Santos, M., Sáez, C., Japón, M. Á., Tortolero, M., Romero, F. G1/S phase progression is regulated by PLK1 degradation through the CDK1/ßTrCP axis.


CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/metabolism , G1 Phase Cell Cycle Checkpoints/physiology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , S Phase Cell Cycle Checkpoints/physiology , beta-Transducin Repeat-Containing Proteins/metabolism , Animals , CDC2 Protein Kinase/genetics , Cell Cycle Proteins/genetics , Cell Line , Cloning, Molecular , Gene Expression Regulation, Enzymologic/physiology , Gene Knockdown Techniques , Humans , Plasmids , Point Mutation , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Two-Hybrid System Techniques , beta-Transducin Repeat-Containing Proteins/genetics , Polo-Like Kinase 1
12.
Am J Physiol Cell Physiol ; 312(3): C341-C353, 2017 Mar 01.
Article En | MEDLINE | ID: mdl-28100484

Activating transcription factor 6 (ATF6), a sensor protein located in the endoplasmic reticulum (ER) membrane, is an important factor in the ER stress signaling pathway. ER stress is known to be involved in folliculogenesis, follicular growth, and ovulation; however, the physiological function of ATF6 in mouse granulosa cells remains largely unknown. The aim of this study was to assess the role of ATF6 in mouse granulosa cells with respect to apoptosis, the cell cycle, and steroid hormone production, as well as several key genes related to follicular development, via RNA interference, immunohistochemical staining, real-time quantitative PCR, Western blotting, flow cytometry, terminal deoxynucleotidyltransferase-mediated deoxy-UTP nick end labeling (TUNEL) assay, and ELISA. Immunohistochemical staining revealed that ATF6 was extensively distributed in the granulosa cells of various ovarian follicles and oocytes in adult female mice. FSH or LH treatment significantly increased ATF6 protein levels in mouse granulosa cells. In the meantime, a recombinant plasmid was used to deplete ATF6 successfully using short hairpin RNA-mediated interference technology, which was verified at both the mRNA and protein levels. Flow cytometry and TUNEL assay analysis indicated that ATF6 depletion decreased apoptosis and arrested the S phase of the cell cycle in mouse granulosa cells. Consistent with these results, p53, caspase-3, B cell lymphoma 2 (Bcl-2)-associated X protein, CCAAT-enhancer-binding protein homologous protein, cyclin A1, cyclin B1, and cyclin D2 mRNA expression decreased, whereas Bcl-2 and glucose-regulated protein 78 kDa mRNA expression increased. Interestingly, ATF6 knockdown obviously increased progesterone and estradiol production in mouse granulosa cells. Cytochrome P450 1b1 (Cyp1b1) mRNA levels were downregulated, whereas Cyp11a1, steroidogenic acute regulatory, and Cyp19a1 mRNA levels were upregulated, in keeping with the changes in steroid hormones. Furthermore, ATF6 disruption remarkably increased insulin-like growth factor binding protein4 (Igfbp4) expression and decreased hyaluronan synthase 2 (Has2), prostaglandin-endoperoxide synthase 2 (Ptgs2), and prostaglandin F receptor (Ptgfr) expression in mouse granulosa cells, which are proteins crucial for follicular development. But, after treating with tunicamycin, the levels of Has2, Ptgs2, and Ptgfr increased relatively, whereas Igfbp4 expression decreased. Collectively, these results imply that ATF6, as a key player in ER stress signaling, may regulate apoptosis, the cell cycle, steroid hormone synthesis, and other modulators related to folliculogenesis in mouse granulosa cells, which may indirectly be involved in the development, ovulation, and atresia of ovarian follicles by affecting the physiological function of granulosa cells. The present study extends our understanding and provides new insights into the physiological significance of ATF6, a key signal transducer of ER stress, in ovarian granulosa cells.


Activating Transcription Factor 6/metabolism , Apoptosis/physiology , Gonadal Steroid Hormones/metabolism , Granulosa Cells/cytology , Granulosa Cells/physiology , S Phase Cell Cycle Checkpoints/physiology , Animals , Cells, Cultured , Female , Gene Knockdown Techniques , Mice
13.
Biomed Pharmacother ; 86: 74-80, 2017 Feb.
Article En | MEDLINE | ID: mdl-27939522

Noscapine is an alkaloid present in the latex of Papaver somniferum. It has been known for its anticancer efficacy and lack of severe toxicities to normal tissues. Structural alterations in noscapine core architecture have produced a number of potent analogues of noscapine. Here, we report an unusual activity of a novel noscapine analogue, 9-(4-vinylphenyl)noscapine (VinPhe-Nos) on cancer cells. As we reported earlier, VinPhe-Nos inhibited MDA-MB-231 cell proliferation with an IC50 of 6µM. The present study elucidated a possible antiproliferative mechanism of action of VinPhe-Nos. The noscapinoid significantly inhibited clonogenic propagation of MDA-MB-231 cells. However, unlike the majority of tubulin-binding agents, it did not induce mitotic arrest; instead, it prolonged S-phase. Although prolonged presence of the drug show some disruption of cellular microtubule architecture, it did not affect microtubule recovery after cold-induced depolymerization. VinPhe-Nos, nevertheless, induced acetylation and bundling of microtubules. Our data suggest that rational modification of parent compound can alter its mechanism of action on cell cycle and that VinPhe-Nos can be investigated further as a less-toxic, S-phase-preferred, cytostatic anticancer agent.


Cell Cycle Checkpoints/physiology , Microtubules/physiology , Noscapine/analogs & derivatives , Noscapine/pharmacology , S Phase Cell Cycle Checkpoints/physiology , Acetylation/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Humans , Microtubules/drug effects , S Phase Cell Cycle Checkpoints/drug effects
14.
Korean J Parasitol ; 54(2): 147-54, 2016 Apr.
Article En | MEDLINE | ID: mdl-27180572

Toxoplasma gondii infection induces alteration of the host cell cycle and cell proliferation. These changes are not only seen in directly invaded host cells but also in neighboring cells. We tried to identify whether this alteration can be mediated by exosomes secreted by T. gondii-infected host cells. L6 cells, a rat myoblast cell line, and RH strain of T. gondii were selected for this study. L6 cells were infected with or without T. gondii to isolate exosomes. The cellular growth patterns were identified by cell counting with trypan blue under confocal microscopy, and cell cycle changes were investigated by flow cytometry. L6 cells infected with T. gondii showed decreased proliferation compared to uninfected L6 cells and revealed a tendency to stay at S or G2/M cell phase. The treatment of exosomes isolated from T. gondii-infected cells showed attenuation of cell proliferation and slight enhancement of S phase in L6 cells. The cell cycle alteration was not as obvious as reduction of the cell proliferation by the exosome treatment. These changes were transient and disappeared at 48 hr after the exosome treatment. Microarray analysis and web-based tools indicated that various exosomal miRNAs were crucial for the regulation of target genes related to cell proliferation. Collectively, our study demonstrated that the exosomes originating from T. gondii could change the host cell proliferation and alter the host cell cycle.


Cell Proliferation , Exosomes/metabolism , M Phase Cell Cycle Checkpoints/physiology , MicroRNAs/biosynthesis , S Phase Cell Cycle Checkpoints/physiology , Toxoplasma/metabolism , Toxoplasmosis/pathology , Animals , Cell Line , Flow Cytometry , Host-Parasite Interactions , Rats , Toxoplasma/pathogenicity , Toxoplasmosis/parasitology
15.
Int J Radiat Oncol Biol Phys ; 94(5): 1207-18, 2016 Apr 01.
Article En | MEDLINE | ID: mdl-27026320

PURPOSE: To determine how low-dose ionizing radiation (LDIR) regulates B lympho-proliferation and its molecular mechanism related with Ikaros, transcription factor. METHODS AND MATERIALS: Splenocytes and IM-9 cells were uniformly irradiated with various doses of a (137)Cs γ-source, and cell proliferation was analyzed. To determine the LDIR-specific phosphorylation of Ikaros, immunoprecipitation and Western blot analysis were performed. To investigate the physiologic function of LDIR-mediatied Ikaros phosphorylation, Ikaros mutants at phosphorylation sites were generated, and cell cycle analysis was performed. RESULTS: First, we found that LDIR enhances B lymphoblast proliferation in an Ikaros-dependent manner. Moreover, we found that LDIR elevates the phosphorylation level of Ikaros protein. Interestingly, we showed that CK2 and AKT are involved in LDIR-induced Ikaros phosphorylation and capable of regulating DNA binding activity of Ikaros via specific phosphorylation. Finally, we identified LDIR-specific Ikaros phosphorylation sites at S391/S393 and showed that the Ikaros phosphorylations at these sites control Ikaros's ability to regulate G1/S cell cycle progression. CONCLUSION: Low-dose ionizing radiation specifically phosphorylates Ikaros protein at Ser 391/393 residues to regulate cell cycle progression in B lymphoblast.


B-Lymphocytes/radiation effects , Casein Kinase II/metabolism , G1 Phase Cell Cycle Checkpoints/physiology , Ikaros Transcription Factor/metabolism , Proto-Oncogene Proteins c-akt/physiology , S Phase Cell Cycle Checkpoints/physiology , B-Lymphocytes/cytology , B-Lymphocytes/physiology , Cell Line , Cell Proliferation/radiation effects , DNA/metabolism , Humans , Ikaros Transcription Factor/genetics , Mutation , Phosphorylation/genetics , Phosphorylation/radiation effects , Radiation Dosage , Radiation, Ionizing
16.
Oncogene ; 35(5): 567-76, 2016 Feb 04.
Article En | MEDLINE | ID: mdl-25893301

The main risk factor for skin cancer is ultraviolet (UV) exposure, which causes DNA damage. Cells respond to UV-induced DNA damage by activating the intra-S-phase checkpoint, which prevents replication fork collapse, late origin firing and stabilizes fragile sites. Recently, the 54-kDa multifunctional protein NONO was found to be involved in the non-homologous end-joining DNA repair process and in poly ADP-ribose polymerase 1 activation. Interestingly, NONO is mutated in several tumour types and emerged as a crucial factor underlying both melanoma development and progression. Therefore, we set out to evaluate whether NONO could be involved in the DNA-damage response to UV radiations. We generated NONO-silenced HeLa cell clones and found that lack of NONO decreased cell growth rate. Then, we challenged NONO-silenced cells with exposure to UV radiations and found that NONO-silenced cells, compared with control cells, continued to synthesize DNA, failed to block new origin firing and impaired CHK1S345 phosphorylation showing a defective checkpoint activation. Consistently, NONO is present at the sites of UV-induced DNA damage where it localizes to RAD9 foci. To position NONO in the DNA-damage response cascade, we analysed the loading onto chromatin of various intra-S-phase checkpoint mediators and found that NONO favours the loading of topoisomerase II-binding protein 1 acting upstream of the ATM and Rad3-related kinase activity. Strikingly, re-expression of NONO, through an sh-resistant mRNA, rescued CHK1S345 phosphorylation in NONO-silenced cells. Interestingly, NONO silencing affected cell response to UV radiations also in a melanoma cell line. Overall, our data uncover a new role for NONO in mediating the cellular response to UV-induced DNA damage.


DNA Damage , Nuclear Matrix-Associated Proteins/physiology , Octamer Transcription Factors/physiology , RNA-Binding Proteins/physiology , S Phase Cell Cycle Checkpoints/physiology , S Phase Cell Cycle Checkpoints/radiation effects , DNA/metabolism , DNA Repair , DNA-Binding Proteins , HeLa Cells , Humans , Nuclear Matrix-Associated Proteins/genetics , Nuclear Matrix-Associated Proteins/metabolism , Octamer Transcription Factors/genetics , Octamer Transcription Factors/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , S Phase Cell Cycle Checkpoints/genetics , Transfection , Ultraviolet Rays
17.
PLoS One ; 10(3): e0119865, 2015.
Article En | MEDLINE | ID: mdl-25822823

Tau is a microtubule-associated protein implicated in neurodegenerative tauopathies. Six tau isoforms are generated from a single gene through alternative splicing of exons 2, 3 and 10 in human brain. Differential expression of tau isoforms has been detected in different brain areas, during neurodevelopment and in neurodegenerative disorders. However, the biological significance of different tau isoforms is not clear. Here, we investigated the individual effect of six different isoforms of tau on cell proliferation and the possible mechanisms by transient expression of eGFP-labeled tau isoform plasmid in N2a cells. Our study showed the transfection efficiency was comparable between different isoforms of tau by examining GFP expression. Compared with other isoforms, we found expression of 1N3R-tau significantly inhibited cell proliferation by Cell Counting Kit-8 assay and BrdU incorporation. Flow cytometry analysis further showed expression of 1N3R-tau induced S phase arrest. Compared with the longest isoform of tau, expression of 1N3R-tau induced cyclin E translocation from the nuclei to cytoplasm, while it did not change the level of cell cycle checkpoint proteins. These data indicate that 1N3R-tau inhibits cell proliferation through inducing S phase arrest.


S Phase Cell Cycle Checkpoints/physiology , tau Proteins/genetics , tau Proteins/physiology , Active Transport, Cell Nucleus , Alternative Splicing , Animals , Cell Line , Cell Proliferation/genetics , Cell Proliferation/physiology , Cell Survival , Cyclin E/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Mice , Neurons/metabolism , Neurons/pathology , Protein Isoforms/genetics , Protein Isoforms/physiology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , S Phase Cell Cycle Checkpoints/genetics , Tauopathies/etiology , Tauopathies/pathology , Tauopathies/physiopathology , Transfection
18.
Anim Reprod Sci ; 155: 80-8, 2015 Apr.
Article En | MEDLINE | ID: mdl-25728901

Follistatin (FST), a local regulator of gonadal functions is a powerful inhibitor of follicle stimulating hormone (FSH) secretion. In the present study, the expression of FST was partially silenced at both transcriptional and translational levels by RNAi-Ready pSIREN-RetroQ-ZsGreen Vector mediated recombinant pshRNA vectors in bovine granulosa cells (bGCs). The results showed that transfection with FST-1 and FST-2 vectors significantly down-regulated mRNA and protein expressions of follistatin by 51% (P = 0.0093) and 72% (P = 0.0078) respectively. After down-regulation of FST in bGCs, cell cycle was arrested at S-phase (9.2 ± 0.6 vs 12.5 ± 0.2, P = 0.0055), and apoptosis was significantly (21.3 ± 2.7 vs 13.9 ± 2.5, P = 0.0051) increased. These findings were further verified by down-regulation of protein level of B-cell leukemia/lymphoma 2 (Bcl2, P = 0.0423), and up-regulation of caspase-3 (P = 0.0362), p21 (P = 0.0067) and mRNA levels of Bcl2-associated X protein (Bax, P = 0.041). Knockdown of FST in bGCs significantly increased activin A concentration in culture medium, while level of estradiol (E2) was suppressed without affecting progesterone production. In addition, mRNA levels of all activin receptor subtypes [activin receptor types I (ACRI) and II (ACRIIA and ACRIIB)] and inhibin α-subunit were augmented (P < 0.05) without altering both inhibin ß-subunits. These findings suggest that follistatin may participate in caspase3-dependent apoptosis through Bcl2/Bax gene family in bovine GCs, whereas, activin and its receptors are associated with its regulation. Activin-induced up-regulation of inhibin-α subunit in bGCs seems to be involved in the regulation of steroidogenesis.


Apoptosis/physiology , Cattle/physiology , Estradiol/metabolism , Follistatin/metabolism , RNA Interference , S Phase Cell Cycle Checkpoints/physiology , Activins/metabolism , Animals , Cells, Cultured , Female , Follistatin/genetics , Granulosa Cells/metabolism , Plasmids , Progesterone/metabolism
19.
Int J Biol Sci ; 10(10): 1193-202, 2014.
Article En | MEDLINE | ID: mdl-25516717

SIRT1, the mammalian homolog of yeast Sir2, is a founding member of a family of 7 protein and histone deacetylases that are involved in numerous biological functions. Previous studies revealed that SIRT1 deficiency results in genome instability, which eventually leads to cancer formation, yet the underlying mechanism is unclear. To investigate this, we conducted a proteomics study and found that SIRT1 interacted with many proteins involved in replication fork protection and origin firing. We demonstrated that loss of SIRT1 resulted in increased replication origin firing, asymmetric fork progression, defective intra-S-phase checkpoint, and chromosome damage. Mechanistically, SIRT1 deacetylates and affects the activity of TopBP1, which plays an essential role in DNA replication fork protection and replication origin firing. Our study demonstrated that ectopic over-expression of the deacetylated form of TopBP1 in SIRT1 mutant cells repressed replication origin firing, while the acetylated form of TopBP1 lost this function. Thus, SIRT1 acts upstream of TopBP1 and plays an essential role in maintaining genome stability by modulating DNA replication fork initiation and the intra-S-phase cell cycle checkpoint.


Carrier Proteins/metabolism , Genomic Instability/genetics , Replication Origin/physiology , S Phase Cell Cycle Checkpoints/physiology , Sirtuin 1/metabolism , Acetylation , Animals , Blotting, Western , Bromodeoxyuridine , Cytogenetic Analysis , Genetic Vectors/genetics , HEK293 Cells , Humans , Immunoprecipitation , Lentivirus , Mass Spectrometry , Mice , Mice, Knockout , RNA, Small Interfering/genetics , Sirtuin 1/genetics
20.
Cell Cycle ; 13(15): 2349-58, 2014.
Article En | MEDLINE | ID: mdl-25483186

Keap1 negatively controls the activity of transcription factor Nrf2. This Keap1/Nrf2 pathway plays a critical role in combating oxidative stress. We aimed at determining whether and how Keap1 modulates the cell cycle of replicating hepatocytes during liver regeneration. Two-thirds partial hepatectomy (PH) was performed on wild-type mice and Keap1+/- (Keap1 knockdown) mice. We found that, following PH, Keap1 knockdown resulted in a delay in S-phase entry, disruption of S-phase progression, and loss of mitotic rhythm of replicating hepatocytes. These events are associated with dysregulation of c-Met, EGFR, Akt1, p70S6K, Cyclin A2, and Cyclin B1 in regenerating livers. Astonishingly, normal regenerating livers exhibited the redox fluctuation coupled with hepatocyte cell cycle progression, while keeping Nrf2 quiescent. Keap1 knockdown caused severe disruption in both the redox cycle and the cell cycle of replicating hepatocytes. Thus, we demonstrate that Keap1 is a potent regulator of hepatic redox cycle and hepatocyte cell cycle during liver regeneration.


Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle/physiology , Cytoskeletal Proteins/metabolism , Hepatocytes/cytology , Liver Regeneration/physiology , Liver/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins/metabolism , Cytoskeletal Proteins/genetics , Hepatectomy , Hepatocytes/metabolism , Kelch-Like ECH-Associated Protein 1 , Liver/cytology , Liver/metabolism , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/metabolism , Oxidation-Reduction , S Phase Cell Cycle Checkpoints/physiology , Signal Transduction
...