Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.214
Filtrar
1.
J Transl Med ; 22(1): 441, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38730481

RESUMO

Microtubule targeting agents (MTAs) are commonly prescribed to treat cancers and predominantly kill cancer cells in mitosis. Significantly, some MTA-treated cancer cells escape death in mitosis, exit mitosis and become malignant polyploid giant cancer cells (PGCC). Considering the low number of cancer cells undergoing mitosis in tumor tissues, killing them in interphase may represent a favored antitumor approach. We discovered that ST-401, a mild inhibitor of microtubule (MT) assembly, preferentially kills cancer cells in interphase as opposed to mitosis, a cell death mechanism that avoids the development of PGCC. Single cell RNA sequencing identified mRNA transcripts regulated by ST-401, including mRNAs involved in ribosome and mitochondrial functions. Accordingly, ST-401 induces a transient integrated stress response, reduces energy metabolism, and promotes mitochondria fission. This cell response may underly death in interphase and avoid the development of PGCC. Considering that ST-401 is a brain-penetrant MTA, we validated these results in glioblastoma cell lines and found that ST-401 also reduces energy metabolism and promotes mitochondria fission in GBM sensitive lines. Thus, brain-penetrant mild inhibitors of MT assembly, such as ST-401, that induce death in interphase through a previously unanticipated antitumor mechanism represent a potentially transformative new class of therapeutics for the treatment of GBM.


Assuntos
Morte Celular , Células Gigantes , Interfase , Microtúbulos , Poliploidia , Humanos , Interfase/efeitos dos fármacos , Microtúbulos/metabolismo , Microtúbulos/efeitos dos fármacos , Linhagem Celular Tumoral , Morte Celular/efeitos dos fármacos , Células Gigantes/efeitos dos fármacos , Células Gigantes/metabolismo , Células Gigantes/patologia , Dinâmica Mitocondrial/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Glioblastoma/patologia , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Glioblastoma/genética , Neoplasias/patologia , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/genética , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos
2.
FASEB J ; 35(10): e21923, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34551143

RESUMO

Our recent studies have shown that haspin, a protein kinase imperative for mitosis, is engaged in the interphase progression of HeLa and U2OS cancer cells. In this investigation, we employed the Fucci reporter system and time-lapse imaging to examine the impact of haspin gene silencing on cell cycle progressions at a single-cell level. We found that the loss of haspin induced multiple cell cycle defects. Specifically, the S/G2 duration was greatly prolonged by haspin gene depletion or inhibition in synchronous HeLa cells. Haspin gene depletion in asynchronous HeLa and U2OS cells led to a similarly protracted S/G2 phase, followed by mitotic cell death or postmitotic G1 arrest. In addition, haspin deficiency resulted in robust induction of the p21CIP1/WAF1 checkpoint protein, a target of the p53 activation. Also, co-depleting haspin with either p21 or p53 could rescue U2OS cells from postmitotic G1 arrest and partially restore their proliferation. These results substantiate the haspin's capacity to regulate interphase and mitotic progression, offering a broader antiproliferative potential of haspin loss in cancer cells.


Assuntos
Ciclo Celular , Proliferação de Células , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Neoplasias/patologia , Proteínas Serina-Treonina Quinases/deficiência , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Corantes Fluorescentes , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Fase G2/efeitos dos fármacos , Humanos , Interfase/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/genética , Mitose/efeitos dos fármacos , Neoplasias/genética , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Fase S/efeitos dos fármacos , Tubercidina/análogos & derivados , Tubercidina/farmacologia , Proteína Supressora de Tumor p53/genética , Ubiquitinação , Regulação para Cima/efeitos dos fármacos
3.
Cell Cycle ; 20(12): 1195-1208, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34048314

RESUMO

ATP metabolism during mitosis needs to be coordinated with numerous energy-demanding activities, especially in cancer cells whose metabolic pathways are reprogramed to sustain rapid proliferation in a nutrient-deficient environment. Although strategies targeting the energy metabolic pathways have shown therapeutic efficacy in preclinical cancer models, how normal cells and cancer cells differentially respond to energy shortage is unclear. In this study, using time-lapse microscopy, we found that cancer cells displayed unique mitotic phenotypes in a dose-dependent manner upon decreasing ATP (i.e. energy) supply. When reduction in ATP concentration was moderate, chromosome movements in mitosis were barely affected, while the metaphase-anaphase transition was significantly prolonged due to reduced tension between the sister-kinetochores, which delayed the satisfaction of the spindle assembly checkpoint. Further reduction in ATP concentration led to a decreased level of Aurora-B at the centromere, resulting in increased chromosome mis-segregation after metaphase delay. In contrast to cancer cells, ATP restriction in non-transformed cells induced cell cycle arrest in interphase, rather than causing mitotic defects. In addition, data mining of cancer patient database showed a correlation between signatures of energy production and chromosomal instability possibly resulted from mitotic defects. Together, these results reveal that energy restriction induces differential responses in normal and cancer cells, with chromosome mis-segregation only observed in cancer cells. This points to targeting energy metabolism as a potentially cancer-selective therapeutic strategy.


Assuntos
Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/farmacologia , Segregação de Cromossomos/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Metáfase/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Neoplasias do Colo do Útero/metabolismo , Anáfase/efeitos dos fármacos , Aurora Quinase B/metabolismo , Feminino , Células HeLa , Humanos , Interfase/efeitos dos fármacos , Cinetocoros/metabolismo , Microscopia/métodos , NAD/farmacologia , Fuso Acromático/metabolismo , Imagem com Lapso de Tempo/métodos , Neoplasias do Colo do Útero/patologia
4.
J Cell Biol ; 220(1)2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-33301566

RESUMO

GRASP55 and GRASP65 have been implicated in stacking of Golgi cisternae and lateral linking of stacks within the Golgi ribbon. However, RNAi or gene knockout approaches to dissect their respective roles have often resulted in conflicting conclusions. Here, we gene-edited GRASP55 and/or GRASP65 with a degron tag in human fibroblasts, allowing for induced rapid degradation by the proteasome. We show that acute depletion of either GRASP55 or GRASP65 does not affect the Golgi ribbon, while chronic degradation of GRASP55 disrupts lateral connectivity of the ribbon. Acute double depletion of both GRASPs coincides with the loss of the vesicle tethering proteins GM130, p115, and Golgin-45 from the Golgi and compromises ribbon linking. Furthermore, GRASP55 and/or GRASP65 is not required for maintaining stacks or de novo assembly of stacked cisternae at the end of mitosis. These results demonstrate that both GRASPs are dispensable for Golgi stacking but are involved in maintaining the integrity of the Golgi ribbon together with GM130 and Golgin-45.


Assuntos
Complexo de Golgi/ultraestrutura , Proteínas da Matriz do Complexo de Golgi/metabolismo , Proteólise , Brefeldina A/farmacologia , Linhagem Celular , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/metabolismo , Humanos , Ácidos Indolacéticos/farmacologia , Interfase/efeitos dos fármacos , Nocodazol/farmacologia , Proteólise/efeitos dos fármacos
5.
Nucleic Acids Res ; 48(14): 7844-7855, 2020 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-32652013

RESUMO

The catalytic activity of human AURORA-A kinase (AURKA) regulates mitotic progression, and its frequent overexpression in major forms of epithelial cancer is associated with aneuploidy and carcinogenesis. Here, we report an unexpected, kinase-independent function for AURKA in DNA replication initiation whose inhibition through a class of allosteric inhibitors opens avenues for cancer therapy. We show that genetic depletion of AURKA, or its inhibition by allosteric but not catalytic inhibitors, blocks the G1-S cell cycle transition. A catalytically inactive AURKA mutant suffices to overcome this block. We identify a multiprotein complex between AURKA and the replisome components MCM7, WDHD1 and POLD1 formed during G1, and demonstrate that allosteric but not catalytic inhibitors prevent the chromatin assembly of functional replisomes. Indeed, allosteric but not catalytic AURKA inhibitors sensitize cancer cells to inhibition of the CDC7 kinase subunit of the replication-initiating factor DDK. Thus, our findings define a mechanism essential for replisome assembly during DNA replication initiation that is vulnerable to inhibition as combination therapy in cancer.


Assuntos
Aurora Quinase A/fisiologia , Replicação do DNA , DNA Polimerase Dirigida por DNA/metabolismo , Complexos Multienzimáticos/metabolismo , Regulação Alostérica , Aurora Quinase A/antagonistas & inibidores , Aurora Quinase A/genética , Aurora Quinase A/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/antagonistas & inibidores , Linhagem Celular , Pontos de Checagem da Fase G1 do Ciclo Celular , Células HeLa , Humanos , Interfase/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Origem de Replicação
6.
Am J Chin Med ; 48(3): 719-736, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32349516

RESUMO

Bauhinia championii (Benth.) is one of the commonly used herbs in Taiwan. The stem of this plant has been used to treat epigastria pain and rheumatoid arthritis. However, the antitumor activities of this herb have never been reported. This study aims to investigate the mechanism of anticancer activity of the extracts from B. championii (BC). BC was fractionated with a series of organic solvents, including n-hexane (H), ethyl acetate (EA), 1-butanol (B), and water (W). We first investigated the effects of BC-H, BC-EA, BC-B and BC-W partitioned fraction on cell viability. In HCT 116 colon cancer cell lines, BC-EA showed the highest inhibition of cell viability and changed the morphology of cells. With dose- and time-dependent manners, BC-EA inhibited the proliferation of HCT 116 cells by inducing apoptosis and G0/G1 phase arrest of cell cycle. To determine the underlying mechanisms, down-regulated CDK2, Cyclin D, and Cyclin E and up-regulated p16, p21, and p53 may account for the cell cycle arrest, while the apoptotic effect of BC-EA may attribute to increased intracellular Ca2+, loss of mitochondria membrane potential (ΔΨm), increase of Bax, Bak, puma, and AIF, and decrease of Bcl-2. Furthermore, the inactivation of Ras signaling pathway by BC-EA also contributed to its apoptotic effect on HCT 116. Our study demonstrates that BC-EA not only inhibits cell growth but also induces apoptosis through inhibiting Ras signal pathway and increasing p53 expression levels. We suggest that BC-EA may be a new dietary supplement and a useful tool to search for therapeutic candidates against colon cancer.


Assuntos
Antineoplásicos Fitogênicos , Apoptose/efeitos dos fármacos , Bauhinia/química , Neoplasias do Colo/patologia , Interfase/efeitos dos fármacos , Extratos Vegetais/farmacologia , Apoptose/genética , Ciclina D/genética , Ciclina D/metabolismo , Quinase 2 Dependente de Ciclina/genética , Quinase 2 Dependente de Ciclina/metabolismo , Relação Dose-Resposta a Droga , Humanos , Interfase/genética , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Fatores de Tempo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
7.
Plant Cell Rep ; 39(8): 1013-1028, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32328702

RESUMO

KEY MESSAGE: Induction of biphasic interphase-mitotic cells and PCC is connected with an increased level of metabolism in root meristem cells of Allium cepa. Previous experiments using primary roots of Allium cepa exposed to low concentrations of hydroxyurea have shown that long-term DNA replication stress (DRS) disrupts essential links of the S-M checkpoint mechanism, leading meristem cells either to premature chromosome condensation (PCC) or to a specific form of chromatin condensation, establishing biphasic organization of cell nuclei with both interphase and mitotic domains (IM cells). The present study supplements and extends these observations by describing general conditions under which both abnormal types of M-phase cells may occur. The analysis of root apical meristem (RAM) cell proliferation after prolonged mild DRS indicates that a broad spectrum of inhibitors is capable of generating PCC and IM organization of cell nuclei. These included: 5-aminouracil (5-AU, a thymine antagonist), characterized by the highest efficiency in creating cells with the IM phenotype, aphidicolin (APH), an inhibitor of DNA polymerase α, 5-fluorodeoxyuridine (FUdR), an inhibitor of thymidylate synthetase, methotrexate (MTX), a folic acid analog that inhibits purine and pyrimidine synthesis, and cytosine arabinoside (Ara-C), which inhibits DNA replication by forming cleavage complexes with topoisomerase I. As evidenced using fluorescence-based click chemistry assays, continuous treatment of onion RAM cells with 5-AU is associated with an accelerated dynamics of the DNA replication machinery and significantly enhanced levels of transcription and translation. Furthermore, DRS conditions bring about an intensified production of hydrogen peroxide (H2O2), depletion of reduced glutathione (GSH), and some increase in DNA fragmentation, associated with only a slight increase in apoptosis-like programmed cell death events.


Assuntos
Replicação do DNA/efeitos dos fármacos , Interfase/efeitos dos fármacos , Meristema/citologia , Mitose/efeitos dos fármacos , Cebolas/citologia , Uracila/análogos & derivados , Apoptose/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Dano ao DNA , Fragmentação do DNA/efeitos dos fármacos , Regulação da Expressão Gênica de Plantas/efeitos dos fármacos , Glutationa/metabolismo , Peróxido de Hidrogênio/metabolismo , Cebolas/genética , Biossíntese de Proteínas/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Plântula/efeitos dos fármacos , Plântula/metabolismo , Transcrição Gênica/efeitos dos fármacos , Uracila/farmacologia
8.
J Biol Chem ; 294(16): 6591-6597, 2019 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-30723159

RESUMO

Mesangial expansion underlies diabetic nephropathy, leading to sclerosis and renal failure. The glycosaminoglycan heparin inhibits mesangial cell growth, but the molecular mechanism is unclear. Here, rat mesangial cells (RMCs) were growth-arrested in the G0/G1 phase of cell division, stimulated to divide in normal glucose (5.6 mm) or high glucose (25.6 mm) with or without heparin, and analyzed for glucose uptake. We observed that RMCs entering the G1 phase in normal glucose with or without heparin rapidly cease glucose uptake. RMCs entering G1 in high glucose sustained glucose uptake for the first 3 h, and high-glucose exposure of RMCs only in the first 8 h of G1 induced the formation of an extracellular monocyte-adhesive hyaluronan matrix after cell division was completed. Moreover, a low heparin concentration under high-glucose conditions blocked glucose uptake by 1 h into G1 Of note, glucose transporter 4 (glut4) localized on the RMC surface at G0/G1 and was internalized into G1 cells under normal glucose conditions with or without heparin within 30 min. We also noted that, under high-glucose conditions, glut4 remained on the RMC surface for at least 2 h into G1 and was internalized by 4 h without heparin and within 1 h with heparin. These results provide evidence that the influx of glucose in hyperglycemic dividing RMCs initiates intermediate glucose metabolism, leading to increased cytosolic UDP sugars, and induces abnormal intracellular hyaluronan synthesis during the S phase of cell division.


Assuntos
Mesângio Glomerular/metabolismo , Transportador de Glucose Tipo 4/metabolismo , Glucose/metabolismo , Heparina/farmacologia , Hiperglicemia/metabolismo , Interfase/efeitos dos fármacos , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Matriz Extracelular/metabolismo , Mesângio Glomerular/patologia , Hiperglicemia/patologia , Ratos
9.
Aging Cell ; 18(1): e12851, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30565836

RESUMO

The Ran GTPase regulates nuclear import and export by controlling the assembly state of transport complexes. This involves the direct action of RanGTP, which is generated in the nucleus by the chromatin-associated nucleotide exchange factor, RCC1. Ran interactions with RCC1 contribute to formation of a nuclear:cytoplasmic (N:C) Ran protein gradient in interphase cells. In previous work, we showed that the Ran protein gradient is disrupted in fibroblasts from Hutchinson-Gilford progeria syndrome (HGPS) patients. The Ran gradient disruption in these cells is caused by nuclear membrane association of a mutant form of Lamin A, which induces a global reduction in heterochromatin marked with Histone H3K9me3 and Histone H3K27me3. Here, we have tested the hypothesis that heterochromatin controls the Ran gradient. Chemical inhibition and depletion of the histone methyltransferases (HMTs) G9a and GLP in normal human fibroblasts reduced heterochromatin levels and caused disruption of the Ran gradient, comparable to that observed previously in HGPS fibroblasts. HMT inhibition caused a defect in nuclear localization of TPR, a high molecular weight protein that, owing to its large size, displays a Ran-dependent import defect in HGPS. We reasoned that pathways dependent on nuclear import of large proteins might be compromised in HGPS. We found that nuclear import of ATM requires the Ran gradient, and disruption of the Ran gradient in HGPS causes a defect in generating nuclear γ-H2AX in response to ionizing radiation. Our data suggest a lamina-chromatin-Ran axis is important for nuclear transport regulation and contributes to the DNA damage response.


Assuntos
Cromatina/metabolismo , Dano ao DNA , Lâmina Nuclear/metabolismo , Transdução de Sinais , Proteína ran de Ligação ao GTP/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Azepinas/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Histonas/metabolismo , Humanos , Interfase/efeitos dos fármacos , Lamina Tipo A/metabolismo , Lisina/metabolismo , Metilação/efeitos dos fármacos , Lâmina Nuclear/efeitos dos fármacos , Progéria/patologia , Quinazolinas/farmacologia , Transdução de Sinais/efeitos dos fármacos
10.
Protoplasma ; 256(3): 763-775, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30554374

RESUMO

Seed priming with iron (Fe) and/or zinc (Zn) can overcome the reduced availability of these micronutrients in soils and crops, but suitable dosages should be predetermined. Nucleolus responds to stress, such as cytotoxicity, with alterations perceivable by cytogenetic analyses. This work intends to study how seed priming with Fe and/or Zn affects the nucleolar activity in roots and the total soluble protein content in the flour of bread wheat cv. 'Jordão'. Seven priming treatments with 0 mg L-1 to 8 mg L-1 of Fe and/or Zn were performed. In all treatments, each metaphase cell presented a maximum of six nucleolar organizer regions positively stained with silver nitrate (Ag-NORs). Also, a maximum number of six nucleoli per nucleus were observed in all treatments, except in the hydroprimed seeds (used as control) that showed a maximum of five nucleoli, probably due to nucleolar fusion. Irregular interphases were frequent in treatments with the highest dosage of micronutrients (8 mg L-1 Fe and/or 8 mg L-1 Zn). The nucleolar area reduced (p < 0.001) as the number of nucleoli increased, and it was lower in treatments with a combination of Fe and Zn. However, the combinations of Fe and Zn showed the highest concentrations of total soluble protein (p ≤ 0.001). Although a reduced nucleolar area represents low ribosomal RNA gene transcription and ribosomal production, the significant increase of the number of nucleoli in the seeds primed with Fe and Zn enhanced the total soluble protein content as compared to the hydroprimed seeds (control) probably due to an increase of nucleolar surface-to-volume ratio that improved the protein synthesis. Overall, this work revealed that priming of bread wheat seeds with suited dosages of Fe and Zn can improve the nutritional value of flour, and the nucleolar number, morphology, and area can be useful biomarkers in cytotoxicity studies.


Assuntos
Pão , Nucléolo Celular/metabolismo , Ferro/farmacologia , Proteínas de Plantas/metabolismo , Sementes/metabolismo , Triticum/metabolismo , Zinco/farmacologia , Nucléolo Celular/efeitos dos fármacos , Interfase/efeitos dos fármacos , Meristema/citologia , Meristema/efeitos dos fármacos , Meristema/metabolismo , Solubilidade
11.
Exp Cell Res ; 367(1): 73-80, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29571950

RESUMO

The Golgi apparatus plays roles in cell polarity, directional cell migration, and bipolar spindle assembly, as well as the secretary pathway. In addition, recent studies have suggested the Golgi-dependent control of mitotic entry. We studied the role of the centrosomal kinase Aurora A in maintaining the Golgi apparatus. Knockdown of Aurora A resulted in Golgi dispersal during interphase. Golgi dispersal was also induced by a selective Aurora A inhibitor, MLN8237. Conversely, overexpression of Aurora A led to tightly packed Golgi apparatus during interphase. Knockdown or inhibition of Aurora A had little or no effect on Golgi vesiculation during mitosis. By synchronizing cell division, we studied whether mitosis was required to induce Golgi dispersal during interphase. Aurora A inhibition induced aberrant mitotic spindle and Golgi dispersal only after mitosis. However, the cells treated with the inhibitor MLN8237 at earlier cell cycle stages (wherein the cells remained undivided) had a normal Golgi architecture. Knockdown or inhibition of Aurora A also led to aberrant integrity of centrosome and Golgi apparatus during interphase. These results suggest that Aurora A activity is involved in the maintenance of Golgi architecture and the relationship between the Golgi apparatus and centrosome.


Assuntos
Aurora Quinase A/metabolismo , Complexo de Golgi/enzimologia , Aurora Quinase A/genética , Azepinas/farmacologia , Linhagem Celular , Centrossomo/efeitos dos fármacos , DNA Complementar/genética , DNA Complementar/metabolismo , Técnicas de Silenciamento de Genes , Complexo de Golgi/efeitos dos fármacos , Humanos , Interfase/efeitos dos fármacos , Interfase/fisiologia , Mitose/efeitos dos fármacos , Mitose/fisiologia , Pirimidinas/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transfecção
12.
Oncogene ; 37(2): 231-240, 2018 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-28925395

RESUMO

Highly expressed in cancer protein 1 (Hec1) is a subunit of the kinetochore (KT)-associated Ndc80 complex, which ensures proper segregation of sister chromatids at mitosis by mediating the interaction between KTs and microtubules (MTs). HEC1 mRNA and protein are highly expressed in many malignancies as part of a signature of chromosome instability. These properties render Hec1 a promising molecular target for developing therapeutic drugs that exert their anticancer activities by producing massive chromosome aneuploidy. A virtual screening study aimed at identifying small molecules able to bind at the Hec1-MT interaction domain identified one positive hit compound and two analogs of the hit with high cytotoxic, pro-apoptotic and anti-mitotic activities. The most cytotoxic analog (SM15) was shown to produce chromosome segregation defects in cancer cells by inhibiting the correction of erroneous KT-MT interactions. Live cell imaging of treated cells demonstrated that mitotic arrest and segregation abnormalities lead to cell death through mitotic catastrophe and that cell death occurred also from interphase. Importantly, SM15 was shown to be more effective in inducing apoptotic cell death in cancer cells as compared to normal ones and effectively reduced tumor growth in a mouse xenograft model. Mechanistically, cold-induced MT depolymerization experiments demonstrated a hyper-stabilization of both mitotic and interphase MTs. Molecular dynamics simulations corroborate this finding by showing that SM15 can bind the MT surface independently from Hec1 and acts as a stabilizer of both MTs and KT-MT interactions. Overall, our studies represent a clear proof of principle that MT-Hec1-interacting compounds may represent novel powerful anticancer agents.


Assuntos
Antineoplásicos/farmacologia , Microtúbulos/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Proteínas Nucleares/antagonistas & inibidores , Animais , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Instabilidade Cromossômica/efeitos dos fármacos , Instabilidade Cromossômica/genética , Segregação de Cromossomos/efeitos dos fármacos , Simulação por Computador , Proteínas do Citoesqueleto , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Humanos , Concentração Inibidora 50 , Interfase/efeitos dos fármacos , Cinetocoros/metabolismo , Masculino , Camundongos , Camundongos Nus , Microtúbulos/metabolismo , Mitose/efeitos dos fármacos , Simulação de Acoplamento Molecular , Neoplasias/patologia , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Domínios Proteicos/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
13.
J Biomed Mater Res A ; 106(3): 621-628, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28986950

RESUMO

A goal of current implantology research is to design devices that induce controlled, guided, and rapid healing. Nanoscale structured substrates [e.g., titania nanotubes (TNTs) or carbon nanotubes (CNTs)] dramatically improve the functions of conventional biomaterials. The present investigation evaluated the behavior of osteoblasts cells cultured on smooth and nanostructured substrates, by measuring osteoblasts specific biomarkers [alkaline phosphatase (AP) and total protein] and cells adhesion strength to substrates, followed by semi-empirical modeling to predict the experimental results. Findings were in total agreement with the current state of the art. The proliferation, as well as the AP and total protein levels were higher on the nanostructure phases (TNTs, CNTs) comparing to the smooth ones (plastic and pure titanium). Cells adhesion strength measured was found higher on the nanostructured materials. This coincided with a higher value of proteins which are directly implicated in the process of adherence. Results were accurately predicted through the Viscoelastic Hybrid Interphase Model. A gradual adherence of bone cells to implants using multilayered biomaterials that involve biodegradable polymeric films and a nanoscale modification of titanium surface is suggested to improve performance through an interphase-mediated osteointegration of orthopedic implants. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 621-628, 2018.


Assuntos
Materiais Biocompatíveis/farmacologia , Modelos Biológicos , Osteoblastos/citologia , Idoso , Fosfatase Alcalina/metabolismo , Adesão Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Módulo de Elasticidade , Elasticidade , Humanos , Interfase/efeitos dos fármacos , Pessoa de Meia-Idade , Osteoblastos/efeitos dos fármacos , Osteoblastos/enzimologia , Estresse Mecânico , Titânio/farmacologia
14.
Mol Cell Endocrinol ; 472: 18-25, 2018 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-29183808

RESUMO

Vitamin D is best known for its regulation of calcium homeostasis. Vitamin D exerts its genomic actions via the vitamin D receptor (VDR). As a member of the superfamily of nuclear receptors (NR), the VDR is primarily located within the nucleus of non-dividing cells. We show here that the VDR relocates from the nucleus into the cytoplasm across all stages of cell division in CHO cells. Furthermore, we show that the VDR is transcriptionally inert during cell division. In addition, 1α, 25 dihydroxyvitamin D (1,25(OH)2D3) promotes VDR binding to the nuclear matrix. Finally, we assessed the structural nature of VDR binding to the nuclear matrix. Mutation of the hinge domain reduced VDR's ability to bind to the nuclear matrix and to initiate transcription in response to 1,25(OH)2D3. Taken together, our data suggest that the association between the VDR and the nuclear matrix accounts for the apparent cytosolic distribution as the matrix disperses within the cytoplasm when cells divide. This may also explain the dramatic reduction in VDR mediated transcription during cell division. Our data also confirm that similar to other NRs, the hinge domain of the VDR is responsible for this association.


Assuntos
Mitose/genética , Matriz Nuclear/metabolismo , Receptores de Calcitriol/química , Receptores de Calcitriol/metabolismo , Transcrição Gênica , Animais , Células CHO , Calcitriol/farmacologia , Cricetinae , Cricetulus , Regulação da Expressão Gênica/efeitos dos fármacos , Interfase/efeitos dos fármacos , Mitose/efeitos dos fármacos , Mutação/genética , Matriz Nuclear/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Domínios Proteicos , Ratos , Relação Estrutura-Atividade , Transcrição Gênica/efeitos dos fármacos
15.
Open Biol ; 7(9)2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28903997

RESUMO

How eukaryotic chromosomes are compacted during mitosis has been a leading question in cell biology since the nineteenth century. Non-histone proteins such as condensin complexes contribute to chromosome shaping, but appear not to be necessary for mitotic chromatin compaction. Histone modifications are known to affect chromatin structure. As histones undergo major changes in their post-translational modifications during mitotic entry, we speculated that the spectrum of cell-cycle-specific histone modifications might contribute to chromosome compaction during mitosis. To test this hypothesis, we isolated core histones from interphase and mitotic cells and reconstituted chromatin with them. We used mass spectrometry to show that key post-translational modifications remained intact during our isolation procedure. Light, atomic force and transmission electron microscopy analysis showed that chromatin assembled from mitotic histones has a much greater tendency to aggregate than chromatin assembled from interphase histones, even under low magnesium conditions where interphase chromatin remains as separate beads-on-a-string structures. These observations are consistent with the hypothesis that mitotic chromosome formation is a two-stage process with changes in the spectrum of histone post-translational modifications driving mitotic chromatin compaction, while the action of non-histone proteins such as condensin may then shape the condensed chromosomes into their classic mitotic morphology.


Assuntos
Montagem e Desmontagem da Cromatina/efeitos dos fármacos , Cromatina/metabolismo , Histonas/metabolismo , Linfócitos/metabolismo , Processamento de Proteína Pós-Traducional , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Animais , Cátions Bivalentes , Linhagem Celular Tumoral , Galinhas , Cromatina/efeitos dos fármacos , Cromatina/ultraestrutura , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Histonas/genética , Humanos , Interfase/efeitos dos fármacos , Linfócitos/efeitos dos fármacos , Linfócitos/ultraestrutura , Magnésio/farmacologia , Microscopia de Força Atômica , Mitose/efeitos dos fármacos , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Nocodazol/farmacologia , Fosforilação , Moduladores de Tubulina/farmacologia
16.
Protoplasma ; 254(1): 461-471, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27048177

RESUMO

The parmalean algae possess a siliceous wall and represent the sister lineage of diatoms; they are thought to be a key group for understanding the evolution of diatoms. Diatoms possess well-characterized and unique mitotic structures, but the mitotic apparatus of Parmales is still unknown. We observed the microtubule (MT) array during interphase and mitosis in Triparma laevis using TEM. The interphase cells had four or five centrioles (∼80 nm in length), from which MTs emanated toward the cytoplasm. In prophase, the bundle of MTs arose at an extranuclear site. The position of centrioles with respect to an MT bundle changed during its elongation. Centrioles were observed on the lateral side of a shorter MT bundle (∼590 nm) and on either side of an extended MT bundle (∼700 nm). In metaphase, the spindle consisted of two types of MTs-MT bundle that passed through a cytoplasmic tunnel in the center of the nucleus and single MTs (possibly kinetochore MTs) that extended from the poles into the nucleus. The nuclear envelope disappeared only at the regions where the kinetochore MTs penetrated. In telophase, daughter chromosomes migrated toward opposite poles, and the MT bundle was observed between segregating chromosomes. These observations showed that MT nucleation does not always occur at the periphery of centrioles through cell cycle and that the spindle of T. laevis has a similar configuration to that of diatoms.


Assuntos
Fuso Acromático/metabolismo , Estramenópilas/metabolismo , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Centríolos/efeitos dos fármacos , Centríolos/metabolismo , Interfase/efeitos dos fármacos , Metáfase/efeitos dos fármacos , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Modelos Biológicos , Silício/farmacologia , Fuso Acromático/efeitos dos fármacos , Estramenópilas/citologia , Estramenópilas/ultraestrutura
17.
J Cell Sci ; 129(24): 4480-4495, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27831493

RESUMO

How spatial organization of the genome depends on nuclear shape is unknown, mostly because accurate nuclear size and shape measurement is technically challenging. In large cell populations of the yeast Saccharomyces cerevisiae, we assessed the geometry (size and shape) of nuclei in three dimensions with a resolution of 30 nm. We improved an automated fluorescence localization method by implementing a post-acquisition correction of the spherical microscopic aberration along the z-axis, to detect the three dimensional (3D) positions of nuclear pore complexes (NPCs) in the nuclear envelope. Here, we used a method called NucQuant to accurately estimate the geometry of nuclei in 3D throughout the cell cycle. To increase the robustness of the statistics, we aggregated thousands of detected NPCs from a cell population in a single representation using the nucleolus or the spindle pole body (SPB) as references to align nuclei along the same axis. We could detect asymmetric changes of the nucleus associated with modification of nucleolar size. Stereotypical modification of the nucleus toward the nucleolus further confirmed the asymmetric properties of the nuclear envelope.


Assuntos
Ciclo Celular , Forma do Núcleo Celular , Microscopia Confocal/métodos , Saccharomycetales/citologia , Carbono/farmacologia , Ciclo Celular/efeitos dos fármacos , Forma do Núcleo Celular/efeitos dos fármacos , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Imageamento Tridimensional , Interfase/efeitos dos fármacos , Membrana Nuclear/efeitos dos fármacos , Membrana Nuclear/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Saccharomycetales/efeitos dos fármacos , Saccharomycetales/metabolismo
18.
PLoS One ; 11(10): e0164601, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27727303

RESUMO

To better understand how the cellular response to DNA replication stress is regulated during embryonic development, we and others have established the early C. elegans embryo as a model system to study this important problem. As is the case in most eukaryotic cell types, the replication stress response is controlled by the ATR kinase in early worm embryos. In this report we use RNAi to systematically characterize ATR pathway components for roles in promoting cell cycle delay during a replication stress response, and we find that these genetic requirements vary, depending on the source of stress. We also examine how individual cell types within the embryo respond to replication stress, and we find that the strength of the response, as defined by duration of cell cycle delay, varies dramatically within blastomeres of the early embryo. Our studies shed light on how the replication stress response is managed in the context of embryonic development and show that this pathway is subject to developmental regulation.


Assuntos
Caenorhabditis elegans/genética , Animais , Proteínas Mutadas de Ataxia Telangiectasia/antagonistas & inibidores , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/antagonistas & inibidores , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Quinase 1 do Ponto de Checagem/antagonistas & inibidores , Quinase 1 do Ponto de Checagem/genética , Quinase 1 do Ponto de Checagem/metabolismo , DNA Helicases/antagonistas & inibidores , DNA Helicases/genética , DNA Helicases/metabolismo , Replicação do DNA/efeitos dos fármacos , Replicação do DNA/efeitos da radiação , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/metabolismo , Embrião não Mamífero/efeitos da radiação , Desenvolvimento Embrionário/efeitos dos fármacos , Desenvolvimento Embrionário/efeitos da radiação , Hidroxiureia/toxicidade , Interfase/efeitos dos fármacos , Interfase/efeitos da radiação , Interferência de RNA , Raios Ultravioleta
19.
Int J Biochem Cell Biol ; 70: 39-47, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26556314

RESUMO

The adenomatous polyposis coli (APC) tumor suppressor is multi-functional. APC is known to localize at the centrosome, and in mitotic cells contributes to formation of the mitotic spindle. To test whether APC contributes to nascent microtubule (MT) growth at interphase centrosomes, we employed MT regrowth assays in U2OS cells to measure MT assembly before and after nocodazole treatment and release. We showed that siRNA knockdown of full-length APC delayed both initial MT aster formation and MT elongation/regrowth. In contrast, APC-mutant SW480 cancer cells displayed a defect in MT regrowth that was unaffected by APC knockdown, but which was rescued by reconstitution of full-length APC. Our findings identify APC as a positive regulator of centrosome MT initial assembly and suggest that this process is disrupted by cancer mutations. We confirmed that full-length APC associates with the MT-nucleation factor γ-tubulin, and found that the APC cancer-truncated form (1-1309) also bound to γ-tubulin through APC amino acids 1-453. While binding to γ-tubulin may help target APC to the site of MT nucleation complexes, additional C-terminal sequences of APC are required to stimulate and stabilize MT growth.


Assuntos
Proteína da Polipose Adenomatosa do Colo/genética , Centrossomo/metabolismo , Células Epiteliais/metabolismo , Microtúbulos/metabolismo , Tubulina (Proteína)/metabolismo , Proteína da Polipose Adenomatosa do Colo/antagonistas & inibidores , Proteína da Polipose Adenomatosa do Colo/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Centrossomo/efeitos dos fármacos , Centrossomo/ultraestrutura , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/ultraestrutura , Regulação da Expressão Gênica , Genes Reporter , Teste de Complementação Genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Interfase/efeitos dos fármacos , Microtúbulos/efeitos dos fármacos , Microtúbulos/ultraestrutura , Mitose/efeitos dos fármacos , Mutação , Nocodazol/farmacologia , Ligação Proteica , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Tubulina (Proteína)/genética , Moduladores de Tubulina/farmacologia
20.
J Cell Sci ; 129(1): 191-205, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26644182

RESUMO

PHD1 (also known as EGLN2) belongs to a family of prolyl hydroxylases (PHDs) that are involved in the control of the cellular response to hypoxia. PHD1 is also able to regulate mitotic progression through the regulation of the crucial centrosomal protein Cep192, establishing a link between the oxygen-sensing and the cell cycle machinery. Here, we demonstrate that PHD1 is phosphorylated by CDK2, CDK4 and CDK6 at S130. This phosphorylation fluctuates with the cell cycle and can be induced through oncogenic activation. Functionally, PHD1 phosphorylation leads to increased induction of hypoxia-inducible factor (HIF) protein levels and activity during hypoxia. PHD1 phosphorylation does not alter its intrinsic enzymatic activity, but instead decreases the interaction between PHD1 and HIF1α. Interestingly, although phosphorylation of PHD1 at S130 lowers its activity towards HIF1α, this modification increases the activity of PHD1 towards Cep192. These results establish a mechanism by which cell cycle mediators, such as CDKs, temporally control the activity of PHD1, directly altering the regulation of HIF1α and Cep192.


Assuntos
Quinases Ciclina-Dependentes/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Fosfosserina/metabolismo , Sequência de Aminoácidos , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Meia-Vida , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia/química , Interfase/efeitos dos fármacos , Mitógenos/farmacologia , Dados de Sequência Molecular , Oncogenes , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Especificidade por Substrato/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA