Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 315
Filtrar
1.
Biomed Pharmacother ; 147: 112645, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35051862

RESUMO

Plants are a rich source for bioactive compounds. However, plant extracts can harbor a mixture of bioactive molecules that promote divergent phenotypes and potentially have confounding effects in bioassays. Even with further purification and identification, target deconvolution can be challenging. Corynoline and acetylcorynoline, are phytochemicals that were previously isolated through a screen for compounds able to induce mitotic arrest and polyploidy in oncogene expressing retinal pigment epithelial (RPE) cells. Here, we shed light on the mechanism by which these phytochemicals can attack human cancer cells. Mitotic arrest was coincident to the induction of centrosome amplification and declustering, causing multi-polar spindle formation. Corynoline was demonstrated to have true centrosome declustering activity in a model where A549 cells were chemically induced to have more than a regular complement of centrosomes. Corynoline could inhibit the centrosome clustering required for pseudo-bipolar spindle formation in these cells. The activity of AURKB, but not AURKA or polo-like kinase 4, was diminished by corynoline. It only partially inhibited AURKB, so it may be a partial antagonist or corynoline may work upstream on an unknown regulator of AURKB activity or localization. Nonetheless, corynoline and acetylcorynoline inhibited the viability of a variety of human cancer derived cell lines. These phytochemicals could serve as prototypes for a next-generation analog with improved potency, selectivity or in vivo bioavailability. Such an analog could be useful as a non-toxic component of combination therapies where inhibiting the chromosomal passenger protein complex is desired.


Assuntos
Aurora Quinase B/efeitos dos fármacos , Alcaloides de Berberina/farmacologia , Mitose/efeitos dos fármacos , Compostos Fitoquímicos/farmacologia , Poliploidia , Células A549 , Apoptose/efeitos dos fármacos , Aurora Quinase A/efeitos dos fármacos , Linhagem Celular Tumoral , Centrossomo/efeitos dos fármacos , Humanos
2.
Biomed Pharmacother ; 144: 112292, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34700231

RESUMO

The centrosome is a special organelle in human cells and an organizing unit for microtubules and signaling molecules. In addition, the centrosome is tightly restricted during the cell cycle and forms the basal body of the cilia in ciliated cells. Centrosome abnormality is frequently observed in malignant tumors. The dysregulation of centrosome-associated proteins leads to multipolar mitosis, aneuploidy, and nondirected cell migration, and therefore promotes cancer progression. The overduplication of primary centrosome and the accumulation of chromosome, comprise the majority cause of chromosomal mis-segregation in cancer cells. This review discusses the structure and function of the centrosome and the role of its associated proteins in the progression of solid tumors. We summarized the effects of centrosome amplification abnormalities and other centrosome-related phenotypes on tumors. The mechanism of the delineation of centrosome amplification with tumor malignancy remains to be decided. A better understanding of centrosome abnormality in tumorigenesis may be useful to screen novel therapeutic strategies for the treatment of solid tumors.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Centrossomo/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Neoplasias/metabolismo , Animais , Antineoplásicos/uso terapêutico , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Centrossomo/efeitos dos fármacos , Centrossomo/patologia , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/genética , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/patologia , Transdução de Sinais
3.
Nat Nanotechnol ; 16(10): 1150-1160, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34354264

RESUMO

Although nanomaterials have shown promising biomedical application potential, incomplete understanding of their molecular interactions with biological systems prevents their inclusion into mainstream clinical applications. Here we show that black phosphorus (BP) nanomaterials directly affect the cell cycle's centrosome machinery. BP destabilizes mitotic centrosomes by attenuating the cohesion of pericentriolar material and consequently leads to centrosome fragmentation within mitosis. As a result, BP-treated cells exhibit multipolar spindles and mitotic delay, and ultimately undergo apoptosis. Mechanistically, BP compromises centrosome integrity by deactivating the centrosome kinase polo-like kinase 1 (PLK1). BP directly binds to PLK1, inducing its aggregation, decreasing its cytosolic mobility and eventually restricting its recruitment to centrosomes for activation. With this mechanism, BP nanomaterials show great anticancer potential in tumour xenografted mice. Together, our study reveals a molecular mechanism for the tumoricidal properties of BP and proposes a direction for biomedical application of nanomaterials by exploring their intrinsic bioactivities.


Assuntos
Proteínas de Ciclo Celular/genética , Centrossomo/efeitos dos fármacos , Nanoestruturas/química , Neoplasias/tratamento farmacológico , Fósforo/farmacologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Animais , Apoptose/efeitos dos fármacos , Proteínas de Ciclo Celular/antagonistas & inibidores , Células HeLa , Xenoenxertos , Humanos , Camundongos , Mitose/efeitos dos fármacos , Neoplasias/genética , Neoplasias/patologia , Fósforo/química , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores
4.
Cells ; 10(6)2021 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-34208028

RESUMO

Etoposide (ETO) has been used in treating adrenocortical tumor (ACT) cells. Our previous study showed that ETO inhibits ACT cell growth. In the present study, we show that ETO treatment at IC50 (10 µM) inhibited ACT cell growth by inducing cellular senescence rather than apoptosis. Several markers of cellular senescence, including enlarged nuclei, activated senescence-associated ß-galactosidase activity, elevated levels of p53 and p21, and down-regulation of Lamin B1, were observed. We further found that ETO induced multiple centrosomes. The inhibition of multiple centrosomes accomplished by treating cells with either roscovitine or centrinone or through the overexpression of NR5A1/SF-1 alleviated ETO-induced senescence, suggesting that ETO triggered senescence via multiple centrosomes. Primary cilia also played a role in ETO-induced senescence. In the mechanism, DNA-PK-Chk2 signaling was activated by ETO treatment; inhibition of this signaling cascade alleviated multiple ETO-induced centrosomes and primary cilia followed by reducing cellular senescence. In addition to DNA damage signaling, autophagy was also triggered by ETO treatment for centrosomal events and senescence. Importantly, the inactivation of DNA-PK-Chk2 signaling reduced ETO-triggered autophagy; however, the inhibition of autophagy did not affect DNA-PK-Chk2 activation. Thus, ETO activated the DNA-PK-Chk2 cascade to facilitate autophagy. The activated autophagy further induced multiple centrosomes and primary cilia followed by triggering senescence.


Assuntos
Neoplasias do Córtex Suprarrenal/patologia , Senescência Celular , Centrossomo/fisiologia , Cílios/efeitos dos fármacos , Etoposídeo/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias do Córtex Suprarrenal/tratamento farmacológico , Neoplasias do Córtex Suprarrenal/genética , Neoplasias do Córtex Suprarrenal/metabolismo , Antineoplásicos Fitogênicos/farmacologia , Apoptose , Autofagia , Proliferação de Células , Centrossomo/efeitos dos fármacos , Dano ao DNA , Humanos , Células Tumorais Cultivadas
5.
Apoptosis ; 26(5-6): 248-252, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33870441

RESUMO

Mitosis, under the control of the microtubule-based mitotic spindle, is an attractive target for anti-cancer treatments, as cancer cells undergo frequent and uncontrolled cell divisions. Microtubule targeting agents that disrupt mitosis or single molecule inhibitors of mitotic kinases or microtubule motors kill cancer cells with a high efficacy. These treatments have, nevertheless, severe disadvantages: they also target frequently dividing healthy tissues, such as the haematopoietic system, and they often lose their efficacy due to primary or acquired resistance mechanisms. An alternative target that has emerged in dividing cancer cells is their ability to "cluster" the poles of the mitotic spindle into a bipolar configuration. This mechanism is necessary for the specific survival of cancer cells that tend to form multipolar spindles due to the frequent presence of abnormal centrosome numbers or other spindle defects. Here we discuss the recent development of combinatorial treatments targeting spindle pole clustering that specifically target cancer cells bearing aberrant centrosome numbers and that have the potential to avoid resistance mechanism due their combinatorial nature.


Assuntos
Antineoplásicos/uso terapêutico , Morte Celular/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Polos do Fuso/efeitos dos fármacos , Antineoplásicos/farmacologia , Centrossomo/efeitos dos fármacos , Centrossomo/metabolismo , Combinação de Medicamentos , Sinergismo Farmacológico , Inibidores de Histona Desacetilases/farmacologia , Inibidores de Histona Desacetilases/uso terapêutico , Humanos , Mitose/efeitos dos fármacos , Neoplasias/patologia , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Polos do Fuso/metabolismo
6.
Front Immunol ; 12: 653100, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33828562

RESUMO

Neutrophils (also called polymorphonuclear leukocytes, PMNs) are heterogeneous and can exhibit considerable phenotypic and functional plasticity. In keeping with this, we discovered previously that Helicobacter pylori infection induces N1-like subtype differentiation of human PMNs that is notable for profound nuclear hypersegmentation. Herein, we utilized biochemical approaches and confocal and super-resolution microscopy to gain insight into the underlying molecular mechanisms. Sensitivity to inhibition by nocodazole and taxol indicated that microtubule dynamics were required to induce and sustain hypersegmentation, and super-resolution Stimulated Emission Depletion (STED) imaging demonstrated that microtubules were significantly more abundant and longer in hypersegmented cells. Dynein activity was also required, and enrichment of this motor protein at the nuclear periphery was enhanced following H. pylori infection. In contrast, centrosome splitting did not occur, and lamin B receptor abundance and ER morphology were unchanged. Finally, analysis of STED image stacks using Imaris software revealed that nuclear volume increased markedly prior to the onset of hypersegmentation and that nuclear size was differentially modulated by nocodazole and taxol in the presence and absence of infection. Taken together, our data define a new mechanism of hypersegmentation that is mediated by microtubules and dynein and as such advance understanding of processes that regulate nuclear morphology.


Assuntos
Dineínas/metabolismo , Infecções por Helicobacter/imunologia , Helicobacter pylori/imunologia , Microtúbulos/metabolismo , Neutrófilos/imunologia , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Centrossomo/efeitos dos fármacos , Centrossomo/metabolismo , Infecções por Helicobacter/microbiologia , Humanos , Microscopia Intravital , Neutrófilos/citologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Nocodazol/farmacologia , Paclitaxel/farmacologia , Cultura Primária de Células , Moduladores de Tubulina/farmacologia
7.
Biochem Biophys Res Commun ; 550: 84-91, 2021 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-33689884

RESUMO

The monopolar spindle 1 ((hMps1/TTK) is a serine/threonine kinase that plays an important role in spindle assembly checkpoint signaling. To explore the possible relationship between TTK inhibition and radiosensitivity, we examined whether TTK inhibition influences cellular susceptibility of radiation. And we further revealed its mechanisms. We found that the expression of TTK was obviously higher in liver cancer tissues compared to the normal liver tissues. Kaplan-Meier Plotter demonstrated that patients with low TTK expression levels had a longer overall survival than patients with high TTK expression levels. TTK inhibitor AZ3146 could simulated liver cancer cells to accumulate in the G2/M phase, which ultimately enhances DNA damage with more γ-H2AX foci and more apoptosis and necrosis induced by radiation, which prompted that TTK inhibition sensitized liver cancer cells to radiation. In addition, TTK inhibition altered cell-cycle progression and exacerbated centrosome abnormalities, resulting in enhanced mitotic catastrophe (MC) induced by radiation in a p21-mediated manner. In this study, we present evidences that the TTK inhibitor promotes the radiosensitivity of liver cancer cells through regulating cell cycle in p21-mediated manner in vitro, indicating that TTK inhibitor may be an attractive radiosensitizer for the patients with liver cancer.


Assuntos
Proteínas de Ciclo Celular/antagonistas & inibidores , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/radioterapia , Proteínas Oncogênicas/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/antagonistas & inibidores , Tolerância a Radiação/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Centrossomo/efeitos dos fármacos , Centrossomo/metabolismo , Centrossomo/efeitos da radiação , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/efeitos da radiação , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos da radiação , Histonas/metabolismo , Humanos , Neoplasias Hepáticas/patologia , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem da Fase M do Ciclo Celular/efeitos da radiação , Necrose/tratamento farmacológico , Necrose/radioterapia , Prognóstico , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Análise de Sobrevida
8.
Nat Commun ; 12(1): 1352, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33649340

RESUMO

Local translation allows for a spatial control of gene expression. Here, we use high-throughput smFISH to screen centrosomal protein-coding genes, and we describe 8 human mRNAs accumulating at centrosomes. These mRNAs localize at different stages during cell cycle with a remarkable choreography, indicating a finely regulated translational program at centrosomes. Interestingly, drug treatments and reporter analyses reveal a common translation-dependent localization mechanism requiring the nascent protein. Using ASPM and NUMA1 as models, single mRNA and polysome imaging reveals active movements of endogenous polysomes towards the centrosome at the onset of mitosis, when these mRNAs start localizing. ASPM polysomes associate with microtubules and localize by either motor-driven transport or microtubule pulling. Remarkably, the Drosophila orthologs of the human centrosomal mRNAs also localize to centrosomes and also require translation. These data identify a conserved family of centrosomal mRNAs that localize by active polysome transport mediated by nascent proteins.


Assuntos
Centrossomo/metabolismo , Polirribossomos/metabolismo , Transporte de RNA , Animais , Proteínas de Ciclo Celular/metabolismo , Centrossomo/efeitos dos fármacos , Cicloeximida/farmacologia , Drosophila/genética , Células HeLa , Humanos , Mitose/efeitos dos fármacos , Fases de Leitura Aberta/genética , Polirribossomos/efeitos dos fármacos , Puromicina/farmacologia , Transporte de RNA/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fuso Acromático/efeitos dos fármacos , Fuso Acromático/metabolismo
9.
Mutagenesis ; 36(1): 95-107, 2021 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-33450026

RESUMO

Bulbus of Fritillaria cirrhosa D. Don (BFC), an outstanding antitussive and expectorant herbal drug used in China and many other countries, has potential but less understood genotoxicity. Previously, we have reported that aqueous extract of BFC compromised the spindle assembly checkpoint and cytokinesis in NCM460 cells. Here, we found that one remarkable observation in BFC-treated NCM460 cells was multipolar mitosis, a trait classically compromises the fidelity of chromosome segregation. More detailed investigation revealed that BFC-induced spindle multipolarity in metaphases and ana-telophases in a dose- and time-dependent manner, suggesting BFC-induced multipolar spindle conformation was not transient. The frequency of multipolar metaphase correlated well to that of multipolar ana-telophases, indicating that BFC-induced multipolar metaphases often persisted through anaphase. Unexpectedly, BFC blocked the proliferation of binucleated cells, suggesting spindle multipolarity was not downstream of BFC-induced cytokinesis failure. Exposure of BFC to early mitotic cells, rather than S/G2 cells, contributed greatly to spindle multipolarity, indicating BFC might disrupt centrosome integrity rather than induce centrosome overduplication. The immunofluorescence results showed that the centrosomes were severely fragmented by a short-term treatment of BFC and the extent of centrosome fragmentation in early mitotic cells was larger than this in S/G2 cells. Consistently, several genes (e.g. p53, Rb centrin-2, Plk-4, Plk-1 and Aurora-A) involved in regulating centrosome integrity were significantly deregulated by BFC. Together, our results suggest that BFC causes multipolar spindles primarily by inducing centrosome fragmentation. Coupling these results to our previous observations, we recommend the risk/benefit ratio should be considered in the practical use of BFC.


Assuntos
Centrossomo/metabolismo , Colo/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Fritillaria/química , Mitose , Extratos Vegetais/farmacologia , Fuso Acromático/efeitos dos fármacos , Centrossomo/efeitos dos fármacos , Colo/metabolismo , Células Epiteliais/metabolismo , Humanos
10.
Arch Toxicol ; 95(2): 703-713, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33057863

RESUMO

1-Methylpyrene (1-MP) is a common environmental pollutant and animal carcinogen. After sequential activation by cytochromes P450 and sulfotransferases, it induced gene mutations and micronuclei in mammalian cells. The type of micronuclei formed, entire chromosomes or fragments, was not analysed. In this study, 1-MP and its primary metabolite, 1-hydroxymethylpyrene (1-HMP), were investigated for the induction of centromere-positive and -negative micronuclei in the human hepatoma cell line HepG2 and its derivative C3A, expressing relevant enzymes at higher levels. Under a short-exposure (9 h)/long-recovery regime (2 cell cycles in total), 1-MP and 1-HMP provided negative test results in HepG2 cells. However, they induced micronuclei in C3A cells, the effect being blocked by 1-aminobenzotriazole (inhibitor of cytochromes P450s) and reduced by pentachlorophenol (inhibitor of sulfotransferases). Immunofluorescence staining of centromere protein B in the micronuclei revealed purely clastogenic effects under this regime. Unexpectedly, 1-MP and 1-HMP at concentrations 1/5-1/4 of that required for micronuclei formation led to mitotic arrest and spindle aberrations, as detected by immunofluorescence staining of ß- and γ-tubulin. Following extended exposure (72 h, 2 cell cycles, no recovery), damage to the spindle apparatus and centrosomes was detected at even lower concentrations, with concurrent formation of micronuclei. At low concentrations (1-8 µM 1-MP, 0.25-0.5 µM 1-HMP), the micronuclei induced were unexceptionally centromere-positive. Thus, the chromosome-damaging mechanism of 1-MP was regime and concentration dependent: potently aneugenic under persistent exposure, while clastogenic at higher concentrations following a short-exposure/long-recovery regime. This is a convincing evidence for the existence of metabolic activation-dependent aneugens.


Assuntos
Micronúcleos com Defeito Cromossômico/efeitos dos fármacos , Mitose/efeitos dos fármacos , Pirenos/toxicidade , Ativação Metabólica/efeitos dos fármacos , Aneugênicos/metabolismo , Aneugênicos/toxicidade , Linhagem Celular Tumoral , Proteína B de Centrômero/metabolismo , Centrossomo/efeitos dos fármacos , Células Hep G2 , Humanos , Testes para Micronúcleos , Microscopia de Fluorescência , Mutagênicos , Pirenos/metabolismo , Fuso Acromático/efeitos dos fármacos
11.
Semin Cell Dev Biol ; 110: 113-122, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32409142

RESUMO

Centrosomes are the major microtubule organizing centers in a large number of animal cells. They are involved in diverse cellular functions like cell division, migration, sensing and motility. Despite being identified more than 100 years ago, they did not receive much attention until recent discoveries suggesting their association with human diseases. Centrosome-related defects have been observed in several human diseases including cancers, brain disorders and ciliopathies. Researchers in the field are trying to understand the relationship between centrosomes and these diseases. Accordingly, this review provides an overview of the current knowledge regarding the role of centrosomes during ciliogenesis and neural stem cell division. The review primarily focuses on the impairment of centrosome number, organization and functioning leading to a wide range of human diseases. Finally, we discuss the scope of targeting centrosomes for therapeutic purposes.


Assuntos
Centrossomo/metabolismo , Cílios/metabolismo , Ciliopatias/genética , Neoplasias/genética , Proteínas do Tecido Nervoso/genética , Animais , Antineoplásicos/farmacologia , Azepinas/farmacologia , Centrossomo/efeitos dos fármacos , Centrossomo/patologia , Centrossomo/ultraestrutura , Cílios/efeitos dos fármacos , Cílios/patologia , Cílios/ultraestrutura , Ciliopatias/metabolismo , Ciliopatias/patologia , Regulação da Expressão Gênica , Humanos , Cirrose Hepática/genética , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Microcefalia/genética , Microcefalia/metabolismo , Microcefalia/patologia , Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Ftalazinas/farmacologia , Pirimidinas/farmacologia , Degeneração Retiniana/genética , Degeneração Retiniana/metabolismo , Degeneração Retiniana/patologia , Transdução de Sinais
12.
J Cell Physiol ; 236(4): 2706-2724, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32869310

RESUMO

Septins play important roles in regulating development and differentiation. Septin 7 (SEPT7) is a crucial component in orchestrating the septin core complex into highly ordered filamentous structures. Here, we showed that genetic depletion of SEPT7 or treatment with forchlorfenuron (FCF; a compound known to affect septin filament assembly) led to reduced the S phase entry in cell models and zebrafish embryos. In addition to colocalizing with actin filaments, SEPT7 resided in the centrosome, and SEPT7 depletion led to aberrant mitotic spindle pole formation. This mitotic defect was rescued in SEPT7-deficient cells by wild-type SEPT7, suggesting that SEPT7 maintained mitotic spindle poles. In addition, we observed disorganized microtubule nucleation and reduced cell migration with SEPT7 depletion. Furthermore, SEPT7 formed a complex with and maintained the abundance of p150glued , the component of centriole subdistal appendages. Depletion of p150glued resulted in a phenotype reminiscent of SEPT7-deficient cells, and overexpression of p150glued reversed the defective phenotypes. Thus, SEPT7 is a centrosomal protein that maintains proper cell proliferation and microtubule array formation via maintaining the abundance of p150glued .


Assuntos
Proteínas de Ciclo Celular/metabolismo , Centrossomo/metabolismo , Complexo Dinactina/metabolismo , Microtúbulos/metabolismo , Fase S , Septinas/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células , Centrossomo/efeitos dos fármacos , Complexo Dinactina/genética , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Microtúbulos/efeitos dos fármacos , Microtúbulos/genética , Compostos de Fenilureia/farmacologia , Piridinas/farmacologia , Fase S/efeitos dos fármacos , Pontos de Checagem da Fase S do Ciclo Celular , Septinas/genética , Transdução de Sinais , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
13.
Biomed Pharmacother ; 132: 110924, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33128942

RESUMO

Centrosome amplification (CA) is a common feature of human tumors, but it is not clear whether this is a cause or a consequence of cancer. The centrosome amplification observed in tumor cells may be explained by a series of events, such as failure of cell division, dysregulation of centrosome cycle checkpoints, and de novo centriole biogenesis disorder. The formation and progression of breast cancer are characterized by genomic abnormality. The centrosomes in breast cancer cells show characteristic structural aberrations, caused by centrosome amplification, which include: an increase in the number and volume of centrosomes, excessive increase of pericentriolar material (PCM), inappropriate phosphorylation of centrosomal molecular, and centrosome clustering formation induced by the dysregulation of important genes. The mechanism of intracellular centrosome amplification, the impact of which on breast cancer and the latest breast cancer target treatment options for centrosome amplification are exhaustively elaborated in this review.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas de Ciclo Celular/metabolismo , Centrossomo/metabolismo , Animais , Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Proteínas de Ciclo Celular/genética , Centrossomo/efeitos dos fármacos , Centrossomo/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Mitose , Terapia de Alvo Molecular , Via de Sinalização Wnt
14.
Nature ; 585(7825): 447-452, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32908313

RESUMO

Genomic instability is a hallmark of cancer, and has a central role in the initiation and development of breast cancer1,2. The success of poly-ADP ribose polymerase inhibitors in the treatment of breast cancers that are deficient in homologous recombination exemplifies the utility of synthetically lethal genetic interactions in the treatment of breast cancers that are driven by genomic instability3. Given that defects in homologous recombination are present in only a subset of breast cancers, there is a need to identify additional driver mechanisms for genomic instability and targeted strategies to exploit these defects in the treatment of cancer. Here we show that centrosome depletion induces synthetic lethality in cancer cells that contain the 17q23 amplicon, a recurrent copy number aberration that defines about 9% of all primary breast cancer tumours and is associated with high levels of genomic instability4-6. Specifically, inhibition of polo-like kinase 4 (PLK4) using small molecules leads to centrosome depletion, which triggers mitotic catastrophe in cells that exhibit amplicon-directed overexpression of TRIM37. To explain this effect, we identify TRIM37 as a negative regulator of centrosomal pericentriolar material. In 17q23-amplified cells that lack centrosomes, increased levels of TRIM37 block the formation of foci that comprise pericentriolar material-these foci are structures with a microtubule-nucleating capacity that are required for successful cell division in the absence of centrosomes. Finally, we find that the overexpression of TRIM37 causes genomic instability by delaying centrosome maturation and separation at mitotic entry, and thereby increases the frequency of mitotic errors. Collectively, these findings highlight TRIM37-dependent genomic instability as a putative driver event in 17q23-amplified breast cancer and provide a rationale for the use of centrosome-targeting therapeutic agents in treating these cancers.


Assuntos
Neoplasias da Mama/genética , Centrossomo/metabolismo , Centrossomo/patologia , Cromossomos Humanos Par 17/genética , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Antineoplásicos/farmacologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Centrossomo/efeitos dos fármacos , Feminino , Fase G2 , Instabilidade Genômica , Humanos , Mitose/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas com Motivo Tripartido/genética , Ubiquitina-Proteína Ligases/genética
15.
Drug Des Devel Ther ; 14: 2987-3000, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32801639

RESUMO

INTRODUCTION: Human gestational choriocarcinoma, a type of gestational trophoblastic disease, occurs after miscarriage, abortion, ectopic pregnancy, or molar pregnancy. Despite recent advances in the mechanism of anticancer drugs that induce human gestational choriocarcinoma apoptosis or block its growth, new therapeutic approaches are needed to be established. Cordycepin is an active anti-cancer component extracted from Cordyceps sinensis. It prevents cell proliferation both in vitro and in vivo. MATERIALS AND METHODS: Here, we examined cell growth by counting cell numbers, and performing a flow cytometry assay and EdU incorporation assay. Centrosome and cytoskeleton-related structures were observed by immunofluorescence assay. The DNA damage-related signaling was examined by Western blot assay. RESULTS: Here, we showed that cordycepin inhibited human gestational choriocarcinoma cell proliferation and induced cell death. In addition, treatment with cordycepin activated DNA-PK and ERK, thus inducing centrosome amplification and aberrant mitosis. These amplified centrosomes also disrupted microtubule arrays and actin networks, thus leading to defective cell adhesion. Furthermore, cordycepin induced autophagy for triggering cell death. CONCLUSION: Thus, our study demonstrates that cordycepin inhibits cell proliferation and disrupts the cytoskeleton by triggering centrosome amplification.


Assuntos
Antineoplásicos/farmacologia , Centrossomo/efeitos dos fármacos , Coriocarcinoma/tratamento farmacológico , Desoxiadenosinas/farmacologia , Doença Trofoblástica Gestacional/tratamento farmacológico , Homeostase/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Coriocarcinoma/patologia , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Doença Trofoblástica Gestacional/patologia , Humanos , Gravidez
16.
Gene ; 760: 144989, 2020 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-32717307

RESUMO

Kinesin 14 family member KIFC1 is a mitotic kinesin which contains a C-terminal motor domain and plays a vital role for clustering the amplified centrosomes. Overexpression of KIFC1 in prostate cancer (PCa) cells showed resistance to docetaxel (DTX). The present study revealed that small KIFC1 inhibitor AZ82 suppresed the transcription and translation of KIFC1 significantly in PCa cells. AZ82 inhibited the KIFC1 expression both in the cytoplasm and nucleus of PCa cells. Inhibition of KIFC1 by AZ82 caused multipolar mitosis in PCa cells via de-clustering the amplified centrosomes and decreased the rate of cancer cell growth and proliferation. Moreover, depletion of KIFC1 reduced cells entering the cell cycle and caused PCa cells death through apoptosis by increasing the expression of Bax and Cytochrome C. Thereby, KIFC1 silencing and inhibition decreased the PCa cells survival by inducing multipolar mitosis as well as apoptosis, suggesting inhibition of KIFC1 using AZ82 might be a strategy to treat PCa by controlling the cancer cell proliferation.


Assuntos
Alanina/análogos & derivados , Centrossomo/efeitos dos fármacos , Cinesinas/antagonistas & inibidores , Neoplasias da Próstata/tratamento farmacológico , Piridinas/farmacologia , Alanina/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Centrossomo/metabolismo , Dineínas/metabolismo , Humanos , Cinesinas/genética , Cinesinas/metabolismo , Masculino , Mitose/efeitos dos fármacos , Miosinas/metabolismo , Próstata/patologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia
17.
J Cell Biol ; 219(4)2020 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-32050025

RESUMO

Centrosomes must resist microtubule-mediated forces for mitotic chromosome segregation. During mitotic exit, however, centrosomes are deformed and fractured by those same forces, which is a key step in centrosome disassembly. How the functional material properties of centrosomes change throughout the cell cycle, and how they are molecularly tuned, remain unknown. Here, we used optically induced flow perturbations to determine the molecular basis of centrosome strength and ductility in C. elegans embryos. We found that both properties declined sharply at anaphase onset, long before natural disassembly. This mechanical transition required PP2A phosphatase and correlated with inactivation of PLK-1 (Polo kinase) and SPD-2 (Cep192). In vitro, PLK-1 and SPD-2 directly protected centrosome scaffolds from force-induced disassembly. Our results suggest that, before anaphase, PLK-1 and SPD-2 respectively confer strength and ductility to the centrosome scaffold so that it can resist microtubule-pulling forces. In anaphase, centrosomes lose PLK-1 and SPD-2 and transition to a weak, brittle state that enables force-mediated centrosome disassembly.


Assuntos
Caenorhabditis elegans/citologia , Centrossomo/metabolismo , Mitose , Animais , Caenorhabditis elegans/efeitos dos fármacos , Caenorhabditis elegans/enzimologia , Caenorhabditis elegans/genética , Centrossomo/efeitos dos fármacos , Leupeptinas/farmacologia , Mitose/efeitos dos fármacos , Mitose/genética
18.
Acta Biochim Biophys Sin (Shanghai) ; 52(1): 72-83, 2020 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-31844893

RESUMO

Type 2 diabetes increases the risk for cancer. Centrosome amplification can initiate tumorigenesis. We have described that type 2 diabetes increases the centrosome amplification of peripheral blood mononuclear cells, with high glucose, insulin, and palmitic acid as the triggers, which suggests that centrosome amplification is a candidate biological mechanism linking diabetes to cancer. In this study, we aimed to further investigate the signaling pathways of the diabetes-associated centrosome amplification and to examine whether and how resveratrol inhibits the centrosome amplification. The results showed that treatment with high glucose, insulin, and palmitic acid, alone or in combination, could increase the protein levels of phospho-protein kinase C alpha (p-PKCα), phospho-p38 mitogen-activated protein kinases (p-p38), c-myc, and c-jun, as well as the mRNA levels of c-myc and c-jun. PKCα inhibitor could inhibit the treatment-induced increase in the protein levels of p-p38, c-myc, and c-jun. Inhibitor or siRNA of p38 was also able to inhibit the treatment-induced increase in the levels of p-p38, c-myc, and c-jun. Meanwhile, knockdown of c-myc or c-jun did not alter the treatment-induced increase in the phosphorylation of PKCα or p38. Importantly, inhibition of the phosphorylation of PKCα or p38 and knockdown of c-myc or c-jun could attenuate the centrosome amplification. In diabetic mice, the levels of p-PKCα, p-p38, c-myc, and c-jun were all increased in the colon tissues. Interestingly, resveratrol, but not metformin, was able to attenuate the treatment-induced increase in the levels of p-PKCα, p-p38, c-myc, and c-jun, as well as the centrosome amplification. In conclusion, our results suggest that PKCα-p38 to c-myc/c-jun is the signaling pathway of the diabetes-associated centrosome amplification, and resveratrol attenuates the centrosome amplification by inhibiting this signaling pathway.


Assuntos
Centrossomo/efeitos dos fármacos , Diabetes Mellitus Experimental/tratamento farmacológico , Proteína Quinase C-alfa/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Resveratrol/farmacologia , Resveratrol/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Centrossomo/metabolismo , Colo/metabolismo , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/metabolismo , Técnicas de Silenciamento de Genes , Glucose/farmacologia , Células HCT116 , Humanos , Insulina/farmacologia , Camundongos , Ácido Palmítico/farmacologia , Fosforilação/efeitos dos fármacos , Proteína Quinase C-alfa/genética , Proteínas Proto-Oncogênicas c-myc/genética , RNA Interferente Pequeno/genética , Estreptozocina/efeitos adversos , Estreptozocina/farmacologia , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno/genética
19.
Bioorg Med Chem ; 28(1): 115154, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31753800

RESUMO

Although cancer cells often harbor supernumerary centrosomes, they form pseudo-bipolar spindles via centrosome clustering, instead of lethal multipolar spindles, and thus avoid cell death. Kinesin-14 HSET/KIFC1 is a crucial protein involved in centrosome clustering. Accordingly, a compound that targets HSET could potentially inhibit cancer cell proliferation in a targeted manner. Here, we report three natural compounds derived from Solidago altissima that restored the growth of fission yeast cells exhibiting lethal HSET overproduction (positive screening), namely solidagonic acid (SA) (1), kolavenic acid analog (KAA: a stereo isomer at C-9 and C-10 of 6ß-tigloyloxykolavenic acid) (2), and kolavenic acid (KA) (3). All three compounds suppressed fission yeast cell death and enabled reversion of the mitotic spindles from a monopolar to bipolar morphology. Compound 2, which exerted the strongest activity against HSET-overproducing yeast cells, also inhibited centrosome clustering in MDA-MB-231 human breast adenocarcinoma cells, which contained large numbers of supernumerary centrosomes. These natural compounds may be useful as bioprobes in studies of HSET function. Moreover, compound 2 is a prime contender in the development of novel agents for cancer treatment.


Assuntos
Diterpenos/farmacologia , Cinesinas/antagonistas & inibidores , Mitose/efeitos dos fármacos , Schizosaccharomyces/efeitos dos fármacos , Linhagem Celular Tumoral , Centrossomo/efeitos dos fármacos , Diterpenos/síntese química , Diterpenos/química , Relação Dose-Resposta a Droga , Humanos , Cinesinas/biossíntese , Estrutura Molecular , Schizosaccharomyces/crescimento & desenvolvimento , Fuso Acromático/efeitos dos fármacos , Relação Estrutura-Atividade
20.
O.F.I.L ; 30(3): 233-238, 2020. tab, ilus
Artigo em Espanhol | IBECS | ID: ibc-200166

RESUMO

OBJETIVO: Determinar los daños citogenéticos generados por el uso de metrotrexate para tratamiento de artritis reumatoidea. METODOLOGÍA: Nuestra experiencia en el uso del metotrexate para sincronización celular como retardante del ciclo celular en fase "S" y su posterior inhibición por competencia con la bromodeoxiuridina en la obtención de cromosomas con bandas "R" de replicación, nos indujo a realizar este estudio citogenético utilizando linfocitos de sangre periférica cultivados en medio PB-Max durante 72 horas, provenientes de una paciente de 46 años de edad, con diagnóstico médico de artritis reumatoidea, tratada con metotrexate durante un mes. RESULTADOS: Clínicamente la paciente presentó inflamación de las articulaciones de los dedos de las manos y hombro izquierdo, con impedimento para flexionar el dedo anular de la mano derecha, además de dolor que impide el movimiento de las mismas articulaciones, en ocasiones y debido al dolor no se puede levantar sin ayuda de su lecho. El resultado del análisis citogenético convencional, indicó que de 50 células analizadas, 23 (46%) presentaron cariotipo normal; 17 (34%) muestran aneuploidías de los diferentes grupos cromosómicos del cariotipo humano incluido el (X) y 10 (20%) de polimorfonucleares anormales. Se discute la etiología génica del control molecular del ciclo celular, pensando en que los hallazgos mencionados, estarían más direccionados a los ciclos aberrantes de duplicación del centrosoma y del huso mitótico en general, así como a la alteración parcial de la síntesis del ADN y el ARN, causados probablemente por la acción del metotrexate, lo cual se ve reflejado en que los hallazgos aneuploídicos en el cariotipo son al azar, sin comprometer en particular cromosomas de un grupo determinado. CONCLUSIONES: Se sugiere a los médicos tratantes tener en cuenta alguna directriz en el protocolo terapéutico, relacionada con la cronicidad y el monitoreo citogenético durante el tratamiento con este medicamento


OBJECTIVE: To determine the cytogenetic damages generated by the use of methotrexate for the treatment of rheumatoid arthritis. METHODS: Our experience in the use of methotrexate for cell synchronization as a retarder of the "S" phase cell cycle and its subsequent inhibition by competition with bromodeoxyuridine in obtaining chromosomes with "R" bands of replication, induced us to perform this cytogenetic study using peripheral blood lymphocytes cultured in PB-Max medium for 72 hours, from a 46-year-old patient, with a medical diagnosis of rheumatoid arthritis, treated with methotrexate for one month. RESULTS: Clinically the patient presented inflammation articulations of left-hand fingers and left shoulder, with impediment flex of the annular finger right hand, besides she has pain that prevent the movement of the same articulations, in occasions and because of this problem she cannot get up without help. The conventionally cytogenetic analysis, shows that of the 50 analyzed cells, 23 (46%) presents normal karyotype; 17 (34%) presents aneuploidies of the chromosomic different groups including de (X) chromosome, and 10 (20%) of abnormal polymorphonuclear. We discuss the genetic etiology of the biology cellular control, keep in mind that mentioned findings are preferentially directed due to aberrant cycles of centrosome duplication and mitotic spindle in general, as well as the partial disturbance of the ADN and ARN synthesis, probably caused by methotrexate action, this fact that is reflected in the aneuploidy findings in the karyotype are to random, without compromise of any particular chromosome of the determined group.  CONCLUSIONS: It is suggested to the treating doctor should really take implement some rule in the therapeutic protocol of corresponding management, relationed with the chronicity and cytogenetic monitoring during the treatment with this medicine


Assuntos
Humanos , Feminino , Pessoa de Meia-Idade , Artrite Reumatoide/tratamento farmacológico , Metotrexato/efeitos adversos , Metotrexato/uso terapêutico , Antirreumáticos/efeitos adversos , Antirreumáticos/uso terapêutico , Cariótipo Anormal/induzido quimicamente , Centrossomo/efeitos dos fármacos , Centríolos/efeitos dos fármacos , Aberrações Cromossômicas/induzido quimicamente , Análise Citogenética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...