Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Plant Physiol ; 176(1): 418-431, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29146775

RESUMO

TANGLED1 (TAN1) and AUXIN-INDUCED-IN-ROOTS9 (AIR9) are microtubule-binding proteins that localize to the division site in plants. Their function in Arabidopsis (Arabidopsis thaliana) remained unclear because neither tan1 nor air9 single mutants have a strong phenotype. We show that tan1 air9 double mutants have a synthetic phenotype consisting of short, twisted roots with disordered cortical microtubule arrays that are hypersensitive to a microtubule-depolymerizing drug. The tan1 air9 double mutants have significant defects in division plane orientation due to failures in placing the new cell wall at the correct division site. Full-length TAN1 fused to yellow fluorescent protein, TAN1-YFP, and several deletion constructs were transformed into the double mutant to assess which regions of TAN1 are required for its function in root growth, root twisting, and division plane orientation. TAN1-YFP expressed in tan1 air9 significantly rescued the double mutant phenotype in all three respects. Interestingly, TAN1 missing the first 126 amino acids, TAN1-ΔI-YFP, failed to rescue the double mutant phenotype, while TAN1 missing a conserved middle region, TAN1-ΔII-YFP, significantly rescued the mutant phenotype in terms of root growth and division plane orientation but not root twisting. We use the tan1 air9 double mutant to discover new functions for TAN1 and AIR9 during phragmoplast guidance and root morphogenesis.


Assuntos
Proteínas de Arabidopsis/genética , Arabidopsis/citologia , Arabidopsis/genética , Proteínas de Ciclo Celular/genética , Divisão Celular , Proteínas Associadas aos Microtúbulos/genética , Mutação/genética , Arabidopsis/efeitos dos fármacos , Proteínas de Arabidopsis/metabolismo , Benzamidas/farmacologia , Padronização Corporal/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos/metabolismo , Paclitaxel/farmacologia , Fenótipo , Raízes de Plantas/citologia , Raízes de Plantas/efeitos dos fármacos , Raízes de Plantas/crescimento & desenvolvimento , Prófase/efeitos dos fármacos , Proteínas Recombinantes de Fusão/metabolismo
2.
Protoplasma ; 254(1): 167-179, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26781092

RESUMO

Histone acetylation plays a critical role in controlling chromatin structure, and reactive oxygen species (ROS) are involved in cell cycle progression. To study the relationship between histone acetylation and cell cycle progression in plants, sodium butyrate (NaB), a histone deacetylase (HDAC) inhibitor that can cause a significant increase in histone acetylation in both mammal and plant genomes, was applied to treat maize seedlings. The results showed that NaB had significant inhibition effects on different root zones at the tissue level and caused cell cycle arrest at preprophase in the root meristem zones. This effect was accompanied by a dramatic increase in the total level of acetylated lysine 9 on histone H3 (H3K9ac) and acetylated lysine 5 on histone H4 (H4K5ac). The exposure of maize roots in NaB led to a continuous rise of intracellular ROS concentration, accompanied by a higher electrolyte leakage ratio and malondialdehyde (MDA) relative value. The NaB-treated group displayed negative results in both TdT-mediated dUTP nick end labelling (TUNEL) and γ-H2AX immunostaining assays. The expression of topoisomerase genes was reduced after treatment with NaB. These results suggested that NaB increased the levels of H3K9ac and H4K5ac and could cause preprophase arrest accompanied with ROS formation leading to the inhibition of DNA topoisomerase.


Assuntos
Ácido Butírico/farmacologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Histonas/metabolismo , Raízes de Plantas/citologia , Prófase/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Zea mays/citologia , Zea mays/metabolismo , Acetilação/efeitos dos fármacos , Cromatina/metabolismo , Ciclinas/genética , Ciclinas/metabolismo , Dano ao DNA/genética , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/genética , DNA Topoisomerases/genética , DNA Topoisomerases/metabolismo , Eletrólitos/metabolismo , Genes de Plantas , Peróxido de Hidrogênio/metabolismo , Malondialdeído/metabolismo , Meristema/citologia , Meristema/efeitos dos fármacos , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Raízes de Plantas/efeitos dos fármacos , Raízes de Plantas/genética , Raízes de Plantas/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Superóxidos/metabolismo , Zea mays/efeitos dos fármacos , Zea mays/crescimento & desenvolvimento
3.
J Cell Biochem ; 117(6): 1340-51, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26529125

RESUMO

Cell division, a prerequisite for cell proliferation, is a process in which each daughter cell inherits one complete set of chromosomes. The mitotic spindle is a dedicated apparatus for the alignment and segregation of chromosomes. Extracellular signal-regulated kinase (ERK) 1/2 plays crucial roles in cell cycle progression, particularly during M-phase. Although, association with the mitotic spindle has been reported, the precise roles played by ERK in the dynamics of the mitotic spindle and in M-phase progression remain to be elucidated. In this study, we used MEK inhibitors U0126 and GSK1120212 to dissect the roles of ERK in M-phase progression and chromosome alignment. Fluorescence microscopy revealed that ERK is localized to the spindle microtubules in a manner independent of Src kinase, which is one of the kinases upstream of ERK at mitotic entry. ERK inhibition induces an increase in the number of prophase cells and a decrease in the number of anaphase cells. Time-lapse imaging revealed that ERK inhibition perturbs chromosome alignment, thereby preventing cells from entering anaphase. These results suggest that ERK plays a role in M-phase progression by regulating chromosome alignment and demonstrate one of the mechanisms by which the aberration of ERK signaling may produce cancer cells.


Assuntos
Cromossomos Humanos/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fuso Acromático/genética , Anáfase/efeitos dos fármacos , Animais , Butadienos/farmacologia , Linhagem Celular , Humanos , Nitrilas/farmacologia , Prófase/efeitos dos fármacos , Piridonas/farmacologia , Pirimidinonas/farmacologia , Fuso Acromático/metabolismo , Suínos , Imagem com Lapso de Tempo
4.
Reproduction ; 151(1): 59-72, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26500283

RESUMO

Binding of 17ß-estradiol (E2) to novel G-protein coupled receptor, Gper1, promotes intra-oocyte adenylyl cyclase activity and transactivates epidermal growth factor receptor to ensure prophase-I arrest. Although involvement of either membrane progestin receptor (mPR) or Igf system has been implicated in regulation of meiosis resumption, possibility of concurrent activation and potential synergism between 17α,20ß-dihydroxy-4-pregnen-3-one (DHP)- and Igf-mediated signalling cascades in alleviating E2 inhibition of oocyte maturation (OM) has not been investigated. Here using zebrafish (Danio rerio) defolliculated oocytes, we examined the effect of DHP and Igf1, either alone or in combination, in presence or absence of E2, on OM in vitro. While priming of denuded oocytes with E2 blocked spontaneous maturation, co-treatment with DHP (3 nM) and Igf1 (10 nM), but not alone, reversed E2 inhibition and promoted a robust increase in germinal vesicle breakdown (GVBD). Although stimulation with either Igf1 or DHP promoted Akt phosphorylation, pharmacological inhibition of PI3K/Akt signalling prevented Igf1-induced GVBD but delayed DHP action till 4-5 h of incubation. Moreover, high intra-oocyte cAMP attenuates both DHP and Igf1-mediated OM and co-stimulation with DHP and Igf1 could effectively reverse E2 action on PKA phosphorylation. Interestingly, data from in vivo studies reveal that heightened expression of igf1, igf3 transcripts in intact follicles corresponded well with elevated phosphorylation of Igf1r and Akt, mPRa immunoreactivity, PKA inhibition and accelerated GVBD response just prior to ovulation. This indicates potential synergism between maturational steroid and Igf1 which might have physiological relevance in overcoming E2 inhibition of meiosis resumption in zebrafish oocytes.


Assuntos
Hidroxiprogesteronas/farmacologia , Oócitos/citologia , Prófase/fisiologia , Somatomedinas/farmacologia , Proteínas de Peixe-Zebra/farmacologia , Peixe-Zebra , Animais , AMP Cíclico/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Sinergismo Farmacológico , Estradiol/farmacologia , Feminino , Oócitos/efeitos dos fármacos , Oócitos/crescimento & desenvolvimento , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Prófase/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Somatomedinas/fisiologia , Proteínas de Peixe-Zebra/fisiologia
5.
Tsitologiia ; 57(6): 459-64, 2015.
Artigo em Russo | MEDLINE | ID: mdl-26495713

RESUMO

We studied cytogenetic polymorphism of the seeds of Pinus sylvestris L. in response to heavy metals exposure in laboratory settings over 2 years' time. We compared results obtained from the seedlings of different years: 2012 and 2013. With an increase in Ni2+ and Cd2+ concentration we observed a decrease in mitotic activity with concurrent rise in the percentage of cells in the prophase. This fact demonstrates the heavy metals act similar to both fixatives and substances that block cleavage spindle formation. In terms of pathological mitosis and the frequency of micronuclei cells, Cd2+ shows higher mutagenity compared to Ni2+. In addition, in the experimental samples, we have distinguished abnormalities such as fragmentations and agglutinations of chromosomes and especially C mitosis occurrence, which are not observed in the control.


Assuntos
Cádmio/farmacologia , Aberrações Cromossômicas/induzido quimicamente , Cromossomos de Plantas/metabolismo , Níquel/farmacologia , Pinus sylvestris/metabolismo , Prófase/efeitos dos fármacos , Cromossomos de Plantas/genética
6.
Plant Signal Behav ; 10(9): e1060385, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26237087

RESUMO

Correct positioning of the division plane is a prerequisite for plant morphogenesis. The preprophase band (PPB) is a key intracellular structure of division site determination. PPB forms in G2 phase as a broad band of microtubules (MTs) that narrows in prophase and specializes few-micrometer-wide cortical belt region, named the cortical division zone (CDZ), in late prophase. The PPB comprises several molecules, some of which act as MT band organization and others remain in the CDZ marking the correct insertion of the cell plate in telophase. Ran GTPase-activating protein (RanGAP) is accumulated in the CDZ and forms a RanGAP band in prophase. However, little is known about when and how RanGAPs gather in the CDZ, and especially with regard to their relationships to MT band formation. Here, we examined the spatial and temporal distribution of RanGAPs and MTs in the preprophase of onion root tip cells using confocal laser scanning microscopy and showed that the RanGAP band appeared in mid-prophase as the width of MT band was reduced to nearly 7 µm. Treatments with cytoskeletal inhibitors for 15 min caused thinning or broadening of the MT band but had little effects on RanGAP band in mid-prophase and most of late prophase cells. Detailed image analyses of the spatial distribution of RanGAP band and MT band showed that the RanGAP band positioned slightly beneath the MT band in mid-prophase. These results raise a possibility that RanGAP behaves differently from MTs during their band formation.


Assuntos
Proteínas Ativadoras de GTPase/metabolismo , Microtúbulos/metabolismo , Proteínas de Plantas/metabolismo , Prófase , Anticorpos/metabolismo , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Dinitrobenzenos/farmacologia , Meristema/citologia , Microtúbulos/efeitos dos fármacos , Modelos Biológicos , Cebolas/citologia , Prófase/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Análise Espaço-Temporal , Sulfanilamidas/farmacologia , Tiazolidinas/farmacologia , Tubulina (Proteína)/metabolismo
7.
PLoS One ; 10(6): e0128551, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26043108

RESUMO

The ankyrin repeat domain 49 (ANKRD49) is an evolutionarily conserved protein highly expressed in testes. However, the function of ANKRD49 in spermatogenesis is unknown. In this study, we found that ANKRD49 resides primarily in nucleus of spermatogonia, spermatocytes and round spermatids. ANKRD49 overexpression augments starvation-induced autophagy in male germ GC-1 cells whereas shRNA knockdown of ANKRD49 attenuates the autophagy. Inhibition of NF-κB pathway by its inhibitors or p65 siRNA prevents the ANKRD49-dependent autophagy augmentation, demonstrating that ANKRD49 enhances autophagy via NF-κB pathway. Our findings suggest that ANKRD49 plays an important role in spermatogenesis via promotion of autophagy-dependent survival.


Assuntos
Repetição de Anquirina , Autofagia , Meios de Cultura Livres de Soro/farmacologia , Proteínas Musculares/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Autofagia/efeitos dos fármacos , Linhagem Celular , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Masculino , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Proteínas Musculares/química , Proteínas Musculares/genética , Prófase/efeitos dos fármacos , Espermatogênese/efeitos dos fármacos , Espermatozoides/efeitos dos fármacos , Espermatozoides/metabolismo , Testículo/efeitos dos fármacos , Testículo/metabolismo , Fatores de Tempo , Transcrição Gênica/efeitos dos fármacos
8.
Cell Cycle ; 13(17): 2674-80, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25486355

RESUMO

In female mice, despite the presence of slight DNA double-strand breaks (DSBs), fully grown oocytes are able to undergo meiosis resumption as indicated by germinal vesicle breakdown (GVBD); however, severe DNA DSBs do reduce and delay entry into M phase through activation of the DNA damage checkpoint. But little is known about the effect of severe DNA DSBs on the spindle assembly checkpoint (SAC) during oocyte maturation. We showed that nearly no first polar body (PB1) was extruded at 12 h of in vitro maturation (IVM) in severe DNA DSBs oocytes, and the limited number of oocytes with PB1 were actually at telophase. However, about 60% of the severe DNA DSBs oocytes which underwent GVBD at 2 h of IVM released a PB1 at 18 h of IVM and these oocytes did reach the second metaphase (MII) stage. Chromosome spread at MI and MII stages showed that chromosomes fragmented after GVBD in severe DNA DSBs oocytes. The delayed PB1 extrusion was due to the disrupted attachment of microtubules to kinetochores and activation of the SAC. At the same time, misaligned chromosome fragments became obvious at the first metaphase (MI) in severe DNA DSBs oocytes. These data implied that the inactivation of SAC during the metaphase-anaphase transition of first meiosis was independent of chromosome integrity. Next, we induced DNA DSBs in vivo, and found that the number of superovulated oocytes per mouse was significantly reduced; moreover, this treatment increased the percentage of apoptotic oocytes. These results suggest that DNA DSBs oocytes undergo apoptosis in vivo.


Assuntos
Linhagem da Célula , Quebras de DNA de Cadeia Dupla , Oócitos/citologia , Oócitos/metabolismo , Animais , Apoptose/efeitos dos fármacos , Bleomicina/farmacologia , Linhagem da Célula/efeitos dos fármacos , Células Cultivadas , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Feminino , Técnicas In Vitro , Cinetocoros/efeitos dos fármacos , Cinetocoros/metabolismo , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Meiose/efeitos dos fármacos , Camundongos Endogâmicos ICR , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Oócitos/efeitos dos fármacos , Corpos Polares/citologia , Corpos Polares/efeitos dos fármacos , Prófase/efeitos dos fármacos , Fatores de Tempo
9.
Biochimie ; 104: 147-55, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24952350

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Prometáfase/efeitos dos fármacos , Pterocarpanos/farmacologia , Antineoplásicos/química , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Prófase/efeitos dos fármacos , Pterocarpanos/química , Fatores de Tempo
10.
Cell Death Dis ; 4: e869, 2013 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-24136233

RESUMO

The G-protein-coupled estrogen receptor 1 (GPER) has recently been reported to mediate the non-genomic action of estrogen in different types of cells and tissues. G-1 (1-[4-(6-bromobenzo[1,3] dioxol-5yl)-3a,4,5,9b-tetrahydro-3H-cyclopenta[c]quinolin-8-yl]-ethanone) was developed as a potent and selective agonist for GPER. G-1 has been shown to induce the expression of genes and activate pathways that facilitate cancer cell proliferation by activating GPER. Here we demonstrate that G-1 has an anticancer potential with a mechanism similar to vinca alkaloids, the commonly used chemotherapy drugs. We found that G-1 blocks tubulin polymerization and thereby interrupts microtubule assembly in ovarian cancer cells leading to the arrest of cell cycle in the prophase of mitosis and the suppression of ovarian cancer cell proliferation. G-1 treatment also induces apoptosis of ovarian cancer cells. The ability of G-1 to target microtubules to suppress ovarian cancer cell proliferation makes it a promising candidate drug for treatment of ovarian cancer.


Assuntos
Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Polimerização/efeitos dos fármacos , Receptores de Estrogênio/agonistas , Receptores Acoplados a Proteínas G/agonistas , Tubulina (Proteína)/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclopentanos/farmacologia , Feminino , Humanos , Prófase/efeitos dos fármacos , Quinolinas/farmacologia , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Fuso Acromático/efeitos dos fármacos , Fuso Acromático/metabolismo , Sus scrofa
11.
Carcinogenesis ; 34(8): 1870-80, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23615397

RESUMO

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.


Assuntos
Glicosídeos Cardíacos/farmacologia , Proteínas de Ciclo Celular/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/genética , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Divisão Celular/efeitos dos fármacos , Divisão Celular/genética , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Fase G2/efeitos dos fármacos , Fase G2/genética , Células HCT116 , Células HT29 , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Microtúbulos/genética , Microtúbulos/metabolismo , NF-kappa B/genética , Prometáfase/efeitos dos fármacos , Prometáfase/genética , Prófase/efeitos dos fármacos , Prófase/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética
12.
Biomaterials ; 34(1): 171-80, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23059005

RESUMO

Anti-angiogenesis is an effective strategy for cancer treatment because uncontrolled tumor growth depends on tumor angiogenesis and sufficient blood supply. Thus, blocking angiogenesis could be a strategy to arrest tumor growth. The function and mechanism of luminescent ruthenium-modified selenium nanoparticles (Ru-SeNPs) in angiogenesis have not been elucidated to date. Here, we found that Ru-SeNPs significantly inhibited human umbilical vascular endothelial cell (HUVEC) proliferation, migration and tube formation. Ru-SeNPs was also tested in vivo in the chicken chorioallantoic membrane (CAM) assay and found to inhibit bFGF-treated CAMs development like suramin. Moreover, we showed that Ru-SeNPs inhibited the activations of FGFR1 and its downstream protein kinases, such ErK and AKT. Furthermore, by using fluorescence confocal microscopy and TEM imaging studies, we have demonstrated their cellular uptake and localization within the cytoplasm of HepG2 and HUVEC cells. These findings indicate that Ru-SeNPs inhibits angiogenesis and may be a viable drug candidate in anti-angiogenesis and anticancer therapies.


Assuntos
Fator 2 de Crescimento de Fibroblastos/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Nanopartículas/química , Neovascularização Fisiológica/efeitos dos fármacos , Piridinas/química , Rutênio/química , Selênio/química , Animais , Anexina A5/metabolismo , Apoptose/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Galinhas , Membrana Corioalantoide/irrigação sanguínea , Membrana Corioalantoide/efeitos dos fármacos , Dicroísmo Circular , Endocitose/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células Hep G2 , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/ultraestrutura , Humanos , Concentração Inibidora 50 , Medições Luminescentes , Nanopartículas/ultraestrutura , Fosforilação/efeitos dos fármacos , Prófase/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Piridinas/síntese química
13.
Biol Res ; 45(2): 177-82, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23096362

RESUMO

We analyzed the in vitro effects of the anti-tumoral drugs doxorubicin, cytosine arabinoside and hydroxyurea on the G2-prophase checkpoint in lymphocytes from healthy individuals. At biologically equivalent concentrations, the induced DNA damage activated the corresponding checkpoint. Thus: i) there was a concentration-dependent delay of G2 time and an increase of both the total DNA lesions produced and repaired before metaphase and; ii) G2-checkpoint adaptation took place as chromosome aberrations (CAs) started to appear in the metaphase, indicating the presence of unrepaired double-strand breaks (DSBs) in the previous G2. The checkpoint ATM/ATR kinases are involved in DSB repair, since the recorded frequency of CAs increased when both kinases were caffeine-abrogated. In genotoxic-treated cells about three-fold higher repair activity was observed in relation to the endogenous background level of DNA lesions. The maximum rate of DNA repaired was 3.4 CAs/100 metaphases/hour, this rise being accompanied by a modest 1.3 fold lengthening of late G2 prophase timing. Because of mitotic chromosome condensation, no DSBs repair can take place until the G1 phase of the next cell cycle, when it occurs by DNA non-homologous end joining (NHEJ). Chromosomal rearrangements formed as a consequence of these error-prone DSB repairs ensure the development of genome instability through the DNA-fusion-bridge cycle. Hence, adaptation of the G2 checkpoint supports the appearance of secondary neoplasia in patients pretreated with genotoxic drugs.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Aberrações Cromossômicas/induzido quimicamente , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Linfócitos/efeitos dos fármacos , Prófase/efeitos dos fármacos , Adulto , Citarabina/toxicidade , Dano ao DNA/efeitos dos fármacos , Doxorrubicina/toxicidade , Feminino , Pontos de Checagem da Fase G2 do Ciclo Celular/genética , Humanos , Hidroxiureia/toxicidade , Linfócitos/citologia , Masculino , Adulto Jovem
14.
Biochem Pharmacol ; 83(9): 1217-28, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22306067

RESUMO

Understanding how colon cancer cells survive within the inflammatory milieu of a tumor, and developing approaches that increase their sensitivity to inflammatory cytokines, may ultimately lead to novel approaches for colon cancer therapy and prevention. Analysis of a number of chemopreventive and therapeutic agents reveal that HDAC inhibitors are particularly adept at sensitizing colon cancer cells TNF or TRAIL mediated apoptosis. In vivo data are consistent with an interaction between SAHA and TNF in inducing apoptosis, as AOM-induced colon tumors express elevated levels of TNF and are more sensitive to SAHA administration. Cell cycle analysis and time-lapse imaging indicated a close correspondence between SAHA-induced prophase arrest and TNF or TRAIL-induced apoptosis. Prophase arrest induced by the Aurora kinase inhibitor VX680 likewise sensitized cells to TNF and TRAIL, with siRNA analysis pointing to Aurora kinase A (and not Aurora kinase B) as being the relevant target for this sensitization. We propose that agents that promote prophase arrest may help sensitize cancer cells to TNF and other inflammatory cytokines. We also discuss how circumvention of an early mitotic checkpoint may facilitate cancer cell survival in the inflammatory micro-environment of the tumor.


Assuntos
Neoplasias do Colo/tratamento farmacológico , Citocinas/farmacologia , Inibidores de Histona Desacetilases/farmacologia , Prófase/efeitos dos fármacos , Animais , Aurora Quinase A , Aurora Quinase B , Aurora Quinases , Butiratos/farmacologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Ensaios de Seleção de Medicamentos Antitumorais , Células HT29 , Humanos , Ácidos Hidroxâmicos/farmacologia , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Vorinostat
15.
Biol. Res ; 45(2): 177-182, 2012. tab
Artigo em Inglês | LILACS | ID: lil-648577

RESUMO

We analyzed the in vitro effects of the anti-tumoral drugs doxorubicin, cytosine arabinoside and hydroxyurea on the G2-prophase checkpoint in lymphocytes from healthy individuals. At biologically equivalent concentrations, the induced DNA damage activated the corresponding checkpoint. Thus: i) there was a concentration-dependent delay of G2 time and an increase of both the total DNA lesions produced and repaired before metaphase and; ii) G2-checkpoint adaptation took place as chromosome aberrations (CAs) started to appear in the metaphase, indicating the presence of unrepaired double-strand breaks (DSBs) in the previous G2. The checkpoint ATM/ATR kinases are involved in DSB repair, since the recorded frequency of CAs increased when both kinases were caffeine-abrogated. In genotoxic-treated cells about three-fold higher repair activity was observed in relation to the endogenous background level of DNA lesions. The maximum rate of DNA repaired was 3.4 CAs/100 metaphases/hour, this rise being accompanied by a modest 1.3 fold lengthening of late G2 prophase timing. Because of mitotic chromosome condensation, no DSBs repair can take place until the G1 phase of the next cell cycle, when it occurs by DNA non-homologous end joining (NHEJ). Chromosomal rearrangements formed as a consequence of these error-prone DSB repairs ensure the development of genome instability through the DNA-fusion-bridge cycle. Hence, adaptation of the G2 checkpoint supports the appearance of secondary neoplasia in patients pretreated with genotoxic drugs.


Assuntos
Adulto , Feminino , Humanos , Masculino , Adulto Jovem , Antibióticos Antineoplásicos/toxicidade , Aberrações Cromossômicas/induzido quimicamente , /efeitos dos fármacos , Linfócitos/efeitos dos fármacos , Prófase/efeitos dos fármacos , Citarabina/toxicidade , Dano ao DNA/efeitos dos fármacos , Doxorrubicina/toxicidade , /genética , Hidroxiureia/toxicidade , Linfócitos/citologia
16.
Mol Plant ; 1(6): 950-60, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19825595

RESUMO

In higher plants, the preprophase band (PPB) of microtubules (MTs) forecasts the cell division site prior to mitosis and specifies the organization of MTs into a bipolar prophase spindle surrounding the nucleus. However, the mechanisms governing this PPB-dependent establishment of bipolarity are unclear. Here, we present evidence from live cell imaging studies that suggest a role for the MTs bridging the PPB and the prophase nucleus in mediating this function. Results from drug treatments, along with genetic evidence from null kinesin plants, suggest that these MTs contribute to the bipolarity, orientation, and position of the prophase spindle. Specifically, the absence of these bridge MTs is associated with lack of bipolarity, while non-uniform distributions of bridge MTs correlate with prophase spindle migration, deformation, and enhanced bipolarity toward the region of highest bridge MT density. This behavior does not require actomyosin-based forces, and is enhanced by suppressing MT dynamics with taxol. These observations occur during late prophase, and are coincident with the gradual closing of annular spindle poles. Based on these data, we describe a hypothetical mechanism for bridge MT-dependent organization of prophase spindles.


Assuntos
/citologia , Prófase , Fuso Acromático/metabolismo , Actinas/metabolismo , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Modelos Biológicos , Paclitaxel/farmacologia , Prófase/efeitos dos fármacos , Fuso Acromático/efeitos dos fármacos , /efeitos dos fármacos
17.
Mol Biol Cell ; 18(10): 3993-4002, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17671163

RESUMO

In higher eukaryotic cells, microtubules within metaphase and anaphase spindles undergo poleward flux, the slow, poleward movement of tubulin subunits through the spindle microtubule lattice. Although a number of studies have documented this phenomenon across a wide range of model systems, the possibility of poleward flux before nuclear envelope breakdown (NEB) has not been examined. Using a mammalian cell line expressing photoactivatable green fluorescent protein (GFP)-tubulin, we observe microtubule motion, both toward and away from centrosomes, at a wide range of rates (0.5-4.5 microm/min) in prophase cells. Rapid microtubule motion in both directions is dynein dependent. In contrast, slow microtubule motion, which occurs at rates consistent with metaphase flux, is insensitive to inhibition of dynein but sensitive to perturbation of Eg5 and Kif2a, two proteins with previously documented roles in flux. Our results demonstrate that microtubules in prophase cells are unexpectedly dynamic and that a subpopulation of these microtubules shows motion that is consistent with flux. We propose that the marked reduction in rate and directionality of microtubule motion from prophase to metaphase results from changes in microtubule organization during spindle formation.


Assuntos
Microtúbulos/metabolismo , Prófase , Animais , Dineínas/metabolismo , Cinesinas/metabolismo , Microtúbulos/efeitos dos fármacos , Movimento/efeitos dos fármacos , Membrana Nuclear/efeitos dos fármacos , Membrana Nuclear/metabolismo , Prometáfase/efeitos dos fármacos , Prófase/efeitos dos fármacos , Pirimidinas/farmacologia , Suínos , Tionas/farmacologia
18.
Oncogene ; 26(45): 6509-17, 2007 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-17471241

RESUMO

A putative ubiquitin conjugating enzyme known as UBE2Q2 was previously identified in a microarray screen for mitotic regulatory proteins. UBE2Q2 is very similar to another human protein, UBE2Q1 and orthologs from other higher eukaryotic species. In these studies, we demonstrate that UBE2Q2 can covalently bind ubiquitin on the active site cysteine in vitro and show that inhibition of this protein in vivo causes an early mitotic arrest and increased cytotoxicity when cells are treated with microtubule inhibiting agents (MIAs). Changes in cell cycle progression and viability are not observed in the absence of MIA treatment, indicating that UBE2Q2 is involved in the response to MIAs rather than performing a more general function in mitosis. Inhibition of the UBE2Q2 protein causes cells to undergo a prolonged prophase arrest suggesting that UBE2Q2 normally functions to antagonize an early mitotic checkpoint. Furthermore, UBE2Q2 inhibition sensitizes cells to the cytotoxic effects of MIAs through caspase-mediated apoptosis that is correlated with PARP-1 cleavage. These data provide insights into the cellular response to MIAs and demonstrate that inhibition of UBE2Q2 protein function may be useful in the treatment of malignancies.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas do Citoesqueleto/fisiologia , Prófase/efeitos dos fármacos , Moduladores de Tubulina/farmacologia , Enzimas de Conjugação de Ubiquitina/fisiologia , Sítios de Ligação , Ciclo Celular , Proteínas do Citoesqueleto/antagonistas & inibidores , Células HeLa , Humanos , Mitose/efeitos dos fármacos , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/metabolismo , Ubiquitina/metabolismo , Enzimas de Conjugação de Ubiquitina/antagonistas & inibidores , Vincristina/farmacologia
19.
Mol Cell Biochem ; 297(1-2): 53-64, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17036164

RESUMO

Creatine kinase (CK) isoenzymes are essential for storing, buffering and intracellular transport of "energy-rich" phosphate compounds in tissues with fluctuating high energy demand such as muscle, brain and other tissues and cells where CK is expressed. In brain and many non-muscle cells, ubiquitous cytosolic "brain-type" BB-CK and ubiquitous mitochondrial CK (uMtCK) act as components of a phosphocreatine shuttle to maintain cellular energy pools and distribute energy flux. To date, still relatively little is known about direct coupling of functional dimeric BB-CK with other partner proteins or enzymes that are important for cell function. Using a global yeast two-hybrid (Y2H) screen with monomeric B-CK as bait and a representative brain cDNA library to search for interaction partners of B-CK with proteins of the brain, we repeatedly identified the cis-Golgi Matrix protein (GM130) as recurrent interacting partner of B-CK. Since HeLa cells also express both BB-CK and GM130, we subsequently used this cellular model system to verify and characterize the BB-CK-GM130 complex by GST-pulldown experiments, as well as by in vivo co-localization studies with confocal microscopy. Using dividing HeLa cells, we report here for the first time that GM130 and BB-CK co-localize specifically in a transient fashion during early prophase of mitosis, when GM130 plays an important role in Golgi fragmentation that starts also at early prophase. These data may shed new light on BB-CK function for energy provision for Golgi-fragmentation that is initiated by cell signalling cascades in the early phases of mitosis.


Assuntos
Autoantígenos/metabolismo , Creatina Quinase Forma BB/metabolismo , Proteínas de Membrana/metabolismo , Prófase , Aminoácidos , Animais , Autoantígenos/química , Brefeldina A/farmacologia , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/metabolismo , Células HeLa , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Proteínas de Membrana/química , Prófase/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Transporte Proteico/efeitos dos fármacos , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Técnicas do Sistema de Duplo-Híbrido
20.
Oncogene ; 24(16): 2589-98, 2005 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-15674323

RESUMO

CHFR, a novel checkpoint gene inactivated in human cancer, delays chromosome condensation in cells treated with microtubule poisons. To understand the molecular mechanism for this delay, we characterized cells with inactivated CHFR and stably transfected derivatives expressing the wild-type gene. After exposure to microtubule poisons, the CHFR-expressing cells arrested transiently in early prophase with a characteristic ruffled morphology of the nuclear envelope and no signs of chromosome condensation. Several markers suggested that Cyclin A/Cdc2 had been activated, whereas Aurora-A and -B and Cyclin B1/Cdc2 were inactive. Further, Cyclin B1 was excluded from the nucleus. Ectopic expression of Cyclin B1 with a mutant nuclear export sequence induced chromosome condensation, and thus overcame the CHFR checkpoint. We conclude that the mechanism by which CHFR delays chromosome condensation involves inhibition of accumulation of Cyclin B1 in the nucleus.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Ciclo Celular/efeitos dos fármacos , Ciclina B/metabolismo , Proteínas de Neoplasias/fisiologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/metabolismo , Ciclina A/metabolismo , Ciclina B1 , Células HCT116 , Humanos , Mitose , Modelos Biológicos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Nocodazol/farmacologia , Proteínas de Ligação a Poli-ADP-Ribose , Prófase/efeitos dos fármacos , Transfecção , Ubiquitina-Proteína Ligases
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...