Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Cell Dev Biol ; 9: 656795, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34026755

RESUMO

The cytolethal distending toxin (CDT) is produced by several Gram-negative pathogenic bacteria. In addition to inflammation, experimental evidences are in favor of a protumoral role of CDT-harboring bacteria such as Escherichia coli, Campylobacter jejuni, or Helicobacter hepaticus. CDT may contribute to cell transformation in vitro and carcinogenesis in mice models, through the genotoxic action of CdtB catalytic subunit. Here, we investigate the mechanism of action by which CDT leads to genetic instability in human cell lines and colorectal organoids from healthy patients' biopsies. We demonstrate that CDT holotoxin induces a replicative stress dependent on CdtB. The slowing down of DNA replication occurs mainly in late S phase, resulting in the expression of fragile sites and important chromosomic aberrations. These DNA abnormalities induced after CDT treatment are responsible for anaphase bridge formation in mitosis and interphase DNA bridge between daughter cells in G1 phase. Moreover, CDT-genotoxic potential preferentially affects human cycling cells compared to quiescent cells. Finally, the toxin induces nuclear distension associated to DNA damage in proliferating cells of human colorectal organoids, resulting in decreased growth. Our findings thus identify CDT as a bacterial virulence factor targeting proliferating cells, such as human colorectal progenitors or stem cells, inducing replicative stress and genetic instability transmitted to daughter cells that may therefore contribute to carcinogenesis. As some CDT-carrying bacterial strains were detected in patients with colorectal cancer, targeting these bacteria could be a promising therapeutic strategy.

2.
Front Cell Dev Biol ; 8: 363, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32582690

RESUMO

Inflammatory Bowel Diseases (IBD) are chronic inflammatory disorders, where epithelial defects drive, at least in part, some of the pathology. We reconstituted human intestinal epithelial organ, by using three-dimension culture of human colon organoids. Our aim was to characterize morphological and functional phenotypes of control (non-IBD) organoids, compared to inflamed organoids from IBD patients. The results generated describe the epithelial defects associated with IBD in primary organoid cultures, and evaluate the use of this model for pharmacological testing of anti-inflammatory approaches. Human colonic tissues were obtained from either surgical resections or biopsies, all harvested in non-inflammatory zones. Crypts were isolated from controls (non-IBD) and IBD patients and were cultured up to 12-days. Morphological (size, budding formation, polarization, luminal content), cell composition (proliferation, differentiation, immaturity markers expression), and functional (chemokine and tight junction protein expression) parameters were measured by immunohistochemistry, RT-qPCR or western-blot. The effects of inflammatory cocktail or anti-inflammatory treatments were studied in controls and IBD organoid cultures respectively. Organoid cultures from controls or IBD patients had the same cell composition after 10 to 12-days of culture, but IBD organoid cultures showed an inflammatory phenotype with decreased size and budding capacity, increased cell death, luminal debris, and inverted polarization. Tight junction proteins were also significantly decreased in IBD organoid cultures. Inflammatory cytokine cocktail reproduced this inflammatory phenotype in non-IBD organoids. Clinically used treatments (5-ASA, glucocorticoids, anti-TNF) reduced some, but not all parameters. Inflammatory phenotype is associated with IBD epithelium, and can be studied in organoid cultures. This model constitutes a reliable human pre-clinical model to investigate new strategies targeting epithelial repair.

4.
Br J Pharmacol ; 175(18): 3656-3668, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29959891

RESUMO

BACKGROUND AND PURPOSE: Thrombin is massively released upon tissue damage associated with bleeding or chronic inflammation. The effects of this thrombin on tissue regrowth and repair has been scarcely addressed and only in cancer cell lines. Hence, the purpose of the present study was to determine thrombin's pharmacological effects on human intestinal epithelium growth, proliferation and apoptosis, using three-dimensional cultures of human colon organoids. EXPERIMENTAL APPROACH: Crypts were isolated from human colonic resections and cultured for 6 days, forming human colon organoids. Cultured organoids were exposed to 10 and 50 mU·mL-1 of thrombin, in the presence or not of protease-activated receptor (PAR) antagonists. Organoid morphology, metabolism, proliferation and apoptosis were followed. KEY RESULTS: Thrombin favoured organoid maturation leading to a decreased number of immature cystic structures and a concomitant increased number of larger structures releasing cell debris and apoptotic cells. The size of budding structures, metabolic activity and proliferation were significantly reduced in organoid cultures exposed to thrombin, while apoptosis was dramatically increased. Both PAR1 and PAR4 antagonists inhibited apoptosis regardless of thrombin doses. Thrombin-induced inhibition of proliferation and metabolic activity were reversed by PAR4 antagonist for thrombin's lowest dose and by PAR1 antagonist for thrombin's highest dose. CONCLUSIONS AND IMPLICATIONS: Overall, our data suggest that the presence of thrombin in the vicinity of human colon epithelial cells favours their maturation at the expense of their regenerative capacities. Our data point to thrombin and its two receptors PAR1 and PAR4 as potential molecular targets for epithelial repair therapies.


Assuntos
Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Colo/efeitos dos fármacos , Organoides/efeitos dos fármacos , Receptor PAR-1/metabolismo , Receptores de Trombina/metabolismo , Trombina/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Colo/citologia , Humanos , Organoides/citologia , Organoides/crescimento & desenvolvimento
5.
Am J Physiol Gastrointest Liver Physiol ; 311(2): G221-36, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27313176

RESUMO

Protease-activated receptors PAR1 and PAR2 play an important role in the control of epithelial cell proliferation and migration. However, the survival of normal and tumor intestinal stem/progenitor cells promoted by proinflammatory mediators may be critical in oncogenesis. The glycogen synthase kinase-3ß (GSK3ß) pathway is overactivated in colon cancer cells and promotes their survival and drug resistance. We thus aimed to determine PAR1 and PAR2 effects on normal and tumor intestinal stem/progenitor cells and whether they involved GSK3ß. First, PAR1 and PAR2 were identified in colon stem/progenitor cells by immunofluorescence. In three-dimensional cultures of murine crypt units or single tumor Caco-2 cells, PAR2 activation decreased numbers and size of normal or cancerous spheroids, and PAR2-deficient spheroids showed increased proliferation, indicating that PAR2 represses proliferation. PAR2-stimulated normal cells were more resistant to stress (serum starvation or spheroid passaging), suggesting prosurvival effects of PAR2 Accordingly, active caspase-3 was strongly increased in PAR2-deficient normal spheroids. PAR2 but not PAR1 triggered GSK3ß activation through serine-9 dephosphorylation in normal and tumor cells. The PAR2-triggered GSK3ß activation implicates an arrestin/PP2A/GSK3ß complex that is dependent on the Rho kinase activity. Loss of PAR2 was associated with high levels of GSK3ß nonactive form, strengthening the role of PAR2 in GSK3ß activation. GSK3 pharmacological inhibition impaired the survival of PAR2-stimulated spheroids and serum-starved cells. Altogether our data identify PAR2/GSK3ß as a novel pathway that plays a critical role in the regulation of stem/progenitor cell survival and proliferation in normal colon crypts and colon cancer.


Assuntos
Colo/enzimologia , Células Epiteliais/enzimologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Células-Tronco Neoplásicas/enzimologia , Receptor PAR-2/metabolismo , Células-Tronco/enzimologia , Animais , Arrestina/metabolismo , Células CACO-2 , Proliferação de Células , Sobrevivência Celular , Colo/patologia , Ativação Enzimática , Células Epiteliais/patologia , Humanos , Masculino , Camundongos Endogâmicos C57BL , Células-Tronco Neoplásicas/patologia , Fosforilação , Proteína Fosfatase 2/metabolismo , Interferência de RNA , Receptor PAR-2/genética , Transdução de Sinais , Esferoides Celulares , Nicho de Células-Tronco , Células-Tronco/patologia , Transfecção , Microambiente Tumoral , Quinases Associadas a rho/metabolismo
6.
Nucleic Acids Res ; 44(4): 1657-68, 2016 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-26578561

RESUMO

Repair of DNA double-strand breaks occurs in a chromatin context that needs to be modified and remodeled to allow suitable access to the different DNA repair machineries. Of particular importance for the maintenance of genetic stability is the tight control of error-prone pathways, such as the alternative End Joining pathway. Here, we show that the chromatin remodeler p400 ATPase is a brake to the use of alternative End Joining. Using specific intracellular reporter susbstrates we observed that p400 depletion increases the frequency of alternative End Joining events, and generates large deletions following repair of double-strand breaks. This increase of alternative End Joining events is largely dependent on CtIP-mediated resection, indicating that it is probably related to the role of p400 in late steps of homologous recombination. Moreover, p400 depletion leads to the recruitment of poly(ADP) ribose polymerase (PARP) and DNA ligase 3 at DNA double-strand breaks, driving to selective killing by PARP inhibitors. All together these results show that p400 acts as a brake to prevent alternative End Joining-dependent genetic instability and underline its potential value as a clinical marker.


Assuntos
Adenosina Trifosfatases/genética , Montagem e Desmontagem da Cromatina/genética , DNA Helicases/genética , Proteínas de Ligação a DNA/genética , Poli(ADP-Ribose) Polimerases/genética , Cromatina/genética , Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades/genética , Instabilidade Genômica/genética , Recombinação Homóloga/genética , Humanos , Inibidores de Poli(ADP-Ribose) Polimerases/administração & dosagem
7.
Carcinogenesis ; 35(11): 2503-11, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25173885

RESUMO

Gem is a small guanosine triphosphate (GTP)-binding protein within the Ras superfamily, involved in the regulation of voltage-gated calcium channel activity and cytoskeleton reorganization. Gem overexpression leads to stress fiber disruption, actin and cell shape remodeling and neurite elongation in interphase cells. In this study, we show that Gem plays a crucial role in the regulation of cortical actin cytoskeleton that undergoes active remodeling during mitosis. Ectopic expression of Gem leads to cortical actin disruption and spindle mispositioning during metaphase. The regulation of spindle positioning by Gem involves its downstream effector Gmip. Knockdown of Gmip rescued Gem-induced spindle phenotype, although both Gem and Gmip accumulated at the cell cortex. In addition, we implicated RhoA GTPase as an important effector of Gem/Gmip signaling. Inactivation of RhoA by overexpressing dominant-negative mutant prevented normal spindle positioning. Introduction of active RhoA rescued the actin and spindle positioning defects caused by Gem or Gmip overexpression. These findings demonstrate a new role of Gem/Gmip/RhoA signaling in cortical actin regulation during early mitotic stages.


Assuntos
Citoesqueleto de Actina/metabolismo , GTP Fosfo-Hidrolases/biossíntese , Proteínas Ativadoras de GTPase/biossíntese , Proteína rhoA de Ligação ao GTP/biossíntese , Citoesqueleto de Actina/genética , GTP Fosfo-Hidrolases/metabolismo , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Mitose/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Transdução de Sinais/genética , Fuso Acromático/genética , Proteína rhoA de Ligação ao GTP/genética
8.
Hum Mol Genet ; 23(8): 2120-31, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24287617

RESUMO

Cancer progression is associated with epigenetic alterations, such as changes in DNA methylation, histone modifications or variants incorporation. The p400 ATPase, which can incorporate the H2A.Z variant, and the Tip60 histone acetyltransferase are interacting chromatin-modifying proteins crucial for the control of cell proliferation. We demonstrate here that Tip60 acts as a tumor suppressor in colon, since mice heterozygous for Tip60 are more susceptible to chemically induced preneoplastic lesions and adenomas. Strikingly, heterozygosity for p400 reverses the Tip60-dependent formation of preneoplastic lesions, uncovering for the first time pro-oncogenic functions for p400. By genome-wide analysis and using a specific inhibitor in vivo, we demonstrated that these effects are dependent on Wnt signaling which is antagonistically impacted by p400 and Tip60: p400 directly favors the expression of a subset of Wnt-target genes and regulators, whereas Tip60 prevents ß-catenin acetylation and activation. Taken together, our data underline the physiopathological importance of interplays between chromatin-modifying enzymes in the control of cancer-related signaling pathways.


Assuntos
Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Histona Acetiltransferases/fisiologia , Histonas/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/fisiologia , Transativadores/fisiologia , Proteínas Wnt/metabolismo , Acetilação , Animais , Western Blotting , Células Cultivadas , Cromatina/metabolismo , Imunoprecipitação da Cromatina , Neoplasias do Colo/genética , Progressão da Doença , Feminino , Perfilação da Expressão Gênica , Histona Acetiltransferases/metabolismo , Lisina Acetiltransferase 5 , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/patologia , Processamento de Proteína Pós-Traducional , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Wnt/genética , beta Catenina/metabolismo
9.
Cell Cycle ; 13(3): 399-407, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24240188

RESUMO

In mammalian cells, DNA double-strand breaks (DSB) can be repaired by 2 main pathways, homologous recombination (HR) and non-homologous end joining (NHEJ). To give access to DNA damage to the repair machinery the chromatin structure needs to be relaxed, and chromatin modifications play major roles in the control of these processes. Among the chromatin modifications, changes in nucleosome composition can influence DNA damage response as observed with the H2A.Z histone variant in yeast. In mammals, p400, an ATPase of the SWI/SNF family able to incorporate H2A.Z in chromatin, was found to be important for histone ubiquitination and BRCA1 recruitment around DSB or for HR in cooperation with Rad51. Recent data with 293T cells showed that mammalian H2A.Z is recruited to DSBs and is important to control DNA resection, therefore participating both in HR and NHEJ. Here we show that depletion of H2A.Z in the osteosarcoma U2OS cell line and in immortalized human fibroblasts does not change parameters of DNA DSB repair while affecting clonogenic ability and cell cycle distribution. In addition, no recruitment of H2A.Z around DSB can be detected in U2OS cells either after local laser irradiation or by chromatin immunoprecipitation. These data suggest that the role of H2A.Z in DSB repair is not ubiquitous in mammals. In addition, given that important cellular parameters, such as cell viability and cell cycle distribution, are more sensitive to H2A.Z depletion than DNA repair, our results underline the difficulty to investigate the role of versatile factors such as H2A.Z.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA , Histonas/genética , Pontos de Checagem do Ciclo Celular/genética , Linhagem Celular Transformada , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Humanos , Lasers
10.
FASEB J ; 26(12): 5025-34, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22964304

RESUMO

Within the Ras superfamily, Gem is a small GTP-binding protein that plays a role in regulating Ca(2+) channels and cytoskeletal remodeling in interphase cells. Here, we report for the first time that Gem is a spindle-associated protein and is required for proper mitotic progression. Functionally, loss of Gem leads to misaligned chromosomes and prometaphase delay. On the basis of different experimental approaches, we demonstrate that loss of Gem by RNA interference induces spindle elongation, while its enforced expression results in spindle shortening. The spindle length phenotype is generated through deregulation of spindle dynamics on Gem depletion and requires the expression of its downstream effector, the kinesin Kif9. Loss of Kif9 induces spindle abnormalities similar to those observed when Gem expression is repressed by siRNA. We further identify Kif9 as a new regulator of spindle dynamics. Kif9 depletion increases the steady-state levels of spindle α-tubulin by increasing the rate of microtubule polymerization. Overall, this study demonstrates a novel mechanism by which Gem contributes to the mitotic progression by maintaining correct spindle length through the kinesin Kif9.


Assuntos
Segregação de Cromossomos , Cinesinas/metabolismo , Proteínas de Membrana/metabolismo , Mitose , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Fuso Acromático/metabolismo , Linhagem Celular , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Immunoblotting , Cinesinas/genética , Proteínas de Membrana/genética , Microscopia de Fluorescência , Microscopia de Vídeo , Microtúbulos/metabolismo , Proteínas Monoméricas de Ligação ao GTP/genética , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tubulina (Proteína)/metabolismo
11.
Eur J Cell Biol ; 88(2): 91-102, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19004523

RESUMO

Amphiphysins are BIN-amphiphysin-RVS (BAR) domain-containing proteins that influence membrane curvature in sites such as T-tubules in muscular cells, endocytic pits in neuronal as well as non-neuronal cells, and possibly cytoplasmic endosomes. This effect on lipid membranes is fulfilled by diverse amphiphysin 2/BIN1 isoforms, generated by alternative splicing and showing distinct structural and functional properties. In this study, our goal was to characterize the functional role of a ubiquitously expressed amphiphysin 2/BIN1 by the characterization of new molecular partners. We performed a two-hybrid screen with an isoform of amphiphysin 2/BIN1 expressed in HeLa cells. We identified CLIP-170 as an amphiphysin 2/BIN1-interacting molecule. CLIP-170 is a plus-end tracking protein involved in microtubule (MT) stability and recruitment of dynactin. The binding between amphiphysin 2/BIN1 and CLIP-170 is dependent on the N-terminal part of amphiphysin 2 (mostly the BAR domain) and an internal coiled-coil region of CLIP-170. This partnership was confirmed by GST pull-down assay and by co-immunoprecipitation in HeLa cells that express endogenous amphiphysin 2 (mostly isoforms 6, 9 and 10). When overexpressed in HeLa cells, amphiphysin 2/BIN1 leads to the formation of intracellular tubules which can closely align with MTs. After MT depolymerization by nocodazole, amphiphysin 2-stained tubules disappear, and reappear after nocodazole washout. Furthermore, depletion of CLIP-170 by RNAi induced a decrease in the proportion of cells with amphiphysin 2-stained tubules and an increase in the proportion of cells with no tubules. This result suggests the existence of a mechanistic link between the two types of tubules, which is likely to involve the +TIP protein, CLIP-170. Amphiphysin 2/BIN1 may be an anchoring point on membranes for CLIP-170, and consequently for MT. Then, the pushing force of polymerizing MT could help amphiphysin 2/BIN1 in its tubulation potential. We propose that amphiphysin 2/BIN1 participates in the tubulation of traffic intermediates and intracellular organelles first via its intrinsic tubulating potential and second via its ability to bind CLIP-170 and MT.


Assuntos
Membrana Celular/metabolismo , Extensões da Superfície Celular/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Membrana Celular/efeitos dos fármacos , Extensões da Superfície Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Células HeLa , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Proteínas Associadas aos Microtúbulos/química , Microtúbulos/efeitos dos fármacos , Proteínas de Neoplasias/química , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Nocodazol/farmacologia , Ligação Proteica/efeitos dos fármacos , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína , Transporte Proteico/efeitos dos fármacos , Tubulina (Proteína)/metabolismo , Técnicas do Sistema de Duplo-Híbrido
12.
Int J Cancer ; 124(6): 1449-56, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19065668

RESUMO

CDC25 phosphatases are key actors in cyclin-dependent kinases activation whose role is essential at various stages of the cell cycle. CDC25 expression is upregulated in a number of human cancers. CDC25 phosphatases are therefore thought to represent promising novel targets in cancer therapy. Here, we report the identification and the characterization of IRC-083864, an original bis-quinone moiety that is a potent and selective inhibitor of CDC25 phosphatases in the low nanomolar range. IRC-083864 inhibits cell proliferation of a number of cell lines, regardless of their resistance to other drugs. It irreversibly inhibits cell proliferation and cell cycle progression and prevents entry into mitosis. In addition, it inhibits the growth of HCT-116 tumor spheroids with induction of p21 and apoptosis. Finally, IRC-083864 reduced tumor growth in mice with established human prostatic and pancreatic tumor xenografts. This study describes a novel compound, which merits further study as a potential anticancer agent.


Assuntos
Benzotiazóis/uso terapêutico , Benzoxazóis/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Quinonas/uso terapêutico , Fosfatases cdc25/antagonistas & inibidores , Animais , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/enzimologia , Neoplasias do Colo/patologia , Quinases Ciclina-Dependentes/metabolismo , Citometria de Fluxo , Humanos , Camundongos , Camundongos Nus , Transplante Heterólogo
13.
Bioorg Med Chem ; 16(19): 9040-9, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18789703

RESUMO

CDC25 phosphatases are considered as attractive targets for anti-cancer therapy. To date, quinone derivatives are among the most potent inhibitors of CDC25 phosphatase activity. We present in this paper the synthesis and the biological evaluation of new quinolinedione and naphthoquinone derivatives, containing carboxylic or malonic acids groups introduced to mimic the role of the phosphate moieties of Cyclin-Dependent Kinase complexes. The most efficient compounds show inhibitory activity against CDC25B with IC(50) values in the 10 microM range, and are cytotoxic against HeLa cells.


Assuntos
Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Naftoquinonas/farmacologia , Quinolonas/farmacologia , Fosfatases cdc25/antagonistas & inibidores , Antineoplásicos/síntese química , Ácidos Carboxílicos/química , Quinases Ciclina-Dependentes/química , Quinases Ciclina-Dependentes/metabolismo , Inibidores Enzimáticos/síntese química , Células HeLa , Humanos , Concentração Inibidora 50 , Malonatos , Mimetismo Molecular , Naftoquinonas/síntese química , Fosfatos/química , Quinolonas/síntese química , Relação Estrutura-Atividade
14.
Eur J Pharmacol ; 591(1-3): 102-5, 2008 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-18616938

RESUMO

Polo-Kinase 1 (PLK1) is a key cell cycle regulator that is necessary for checkpoint recovery after DNA damage-induced G2 arrest. We have examined the effects of PLK inhibition in Acute Myelocytic Leukaemia (AML) cells, whose resistance to genotoxic agents is thought to be associated with checkpoint reinforcement. We report that in U937 AML cells, PLK1 participates in checkpoint recovery, and that inhibition of PLK by the GW843682X compound results in mitotic accumulation and apoptosis. We also found that when challenged with VP-16, inhibition of PLK1 prevented U937 cells from checkpoint exit. Finally, we found that treatment with GW843682X slightly reduced genotoxic-induced inhibition of colony formation efficiency of primary leukaemia cells (CFU-L) from AML patients.


Assuntos
Benzimidazóis/farmacologia , Proteínas de Ciclo Celular/antagonistas & inibidores , Leucemia Mieloide Aguda/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Tiofenos/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Proteínas de Ciclo Celular/metabolismo , Divisão Celular/efeitos dos fármacos , Etoposídeo/farmacologia , Fase G2/efeitos dos fármacos , Humanos , Mitose/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Células U937 , Quinase 1 Polo-Like
16.
Mol Cancer Ther ; 5(6): 1446-51, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16818502

RESUMO

Cell cycle arrest at the G2-M checkpoint is an essential feature of the mechanisms that preserve genomic integrity. CDC25 phosphatases control cell cycle progression by dephosphorylating and activating cyclin-dependent kinase/cyclin complexes. Their activities are, therefore, tightly regulated to modulate cell cycle arrest in response to DNA damage exposure. Here, we report that overexpression of CDC25B affects viability, reduces clonogenic efficiency, and increases sensitivity of cancer cells to a genotoxic agent. We show that ectopic expression of CDC25B results in bypass of a genotoxic-induced G2-M checkpoint. In addition, cancer cells constitutively expressing high level of CDC25B are shown to be prone to exit prematurely from the G2-M checkpoint arrest and to enter mitosis. Finally, we show that this exit is dependent on CDC25B expression. Together with previous results, our data strongly support a model in which CDC25B is the key phosphatase that controls entry into mitosis after DNA damage, thus emphasizing the relevance of its overexpression in many human tumors.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Fosfatases cdc25/metabolismo , Antineoplásicos Fitogênicos/farmacologia , Western Blotting , Proteínas de Ciclo Celular/genética , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/fisiologia , Etoposídeo/farmacologia , Citometria de Fluxo , Técnica Indireta de Fluorescência para Anticorpo , Fase G2/efeitos dos fármacos , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Tetraciclina/farmacologia , Células Tumorais Cultivadas , Ensaio Tumoral de Célula-Tronco , Fosfatases cdc25/genética
17.
Mol Cancer Ther ; 4(9): 1378-87, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16170030

RESUMO

Cell cycle regulators, such as the CDC25 phosphatases, are potential targets for the development of new anticancer drugs. Here we report the identification and the characterization of BN82685, a quinone-based CDC25 inhibitor that is active in vitro and in vivo. BN82685 inhibits recombinant CDC25A, B, and C phosphatases in vitro. It inhibits the growth of human tumor cell lines with an IC(50) in the submicromolar range, independently of their resistance to chemotherapeutic agents. This inhibitory effect is irreversible on both the purified CDC25 enzyme in vitro and on tumor cell proliferation. The specificity of BN82685 towards the CDC25 phosphatases is shown by an increase in cyclin-dependent kinase 1 tyrosine 15 phosphorylation, by the reversion of the mitosis-inducing effect of CDC25B overexpression in HeLa cells, and by the lack of a growth inhibitory effect in an assay based on the use of a CDC25-independent fission yeast model. Finally, when administered p.o., BN82685 is shown to inhibit the growth of the human pancreatic tumor Mia PaCa-2 xenografted in athymic nude mice. BN82685 is therefore a promising new compound targeting CDC25, which confirms the interest of the inhibition of these enzymes as an anticancer therapeutic strategy.


Assuntos
Benzoquinonas/farmacologia , Inibidores Enzimáticos/farmacologia , Neoplasias Pancreáticas/patologia , Tiazóis/farmacologia , Fosfatases cdc25/antagonistas & inibidores , Administração Oral , Animais , Benzoquinonas/administração & dosagem , Benzoquinonas/síntese química , Disponibilidade Biológica , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Células HeLa , Humanos , Camundongos , Camundongos Nus , Mitose/efeitos dos fármacos , Neoplasias Pancreáticas/enzimologia , Schizosaccharomyces/genética , Schizosaccharomyces/crescimento & desenvolvimento , Schizosaccharomyces/metabolismo , Tiazóis/administração & dosagem , Tiazóis/síntese química , Transplante Heterólogo , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Bioorg Med Chem ; 13(16): 4871-9, 2005 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15921913

RESUMO

CDC25 dual-specificity phosphatases are essential key regulators of eukaryotic cell cycle progression and the CDC25A and B isoforms are over-expressed in different tumors and related cancer cell lines. CDC25s are now considered to be interesting targets in the search for novel anticancer agents. We describe new compounds derived from vitamin K3 that inhibit CDC25B activity with IC50 values in the low micromolar range. These naphthoquinone derivatives also display antiproliferative activity on HeLa cells as expected for CDC25 inhibitors and inhibit cell growth in a clonogenic assay at submicromolar concentrations. They increase inhibitory tyrosine 15 phosphorylation of CDK and induce the cleavage of PARP, a hallmark of apoptosis.


Assuntos
Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Naftoquinonas/síntese química , Naftoquinonas/farmacologia , Fosfatases cdc25/antagonistas & inibidores , Antineoplásicos/síntese química , Antineoplásicos/química , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/antagonistas & inibidores , Linhagem Celular Tumoral , Desenho de Fármacos , Inibidores Enzimáticos/química , Células HeLa , Humanos , Concentração Inibidora 50 , Modelos Moleculares , Naftoquinonas/química , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Vitamina K 3/análogos & derivados , Vitamina K 3/farmacologia , Fosfatases cdc25/química , Fosfatases cdc25/metabolismo
19.
Cell Cycle ; 4(6): 806-11, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15908796

RESUMO

The phosphatase CDC25B is one of the key regulators that control entry into mitosis through the dephosphorylation and subsequent activation of the cyclin-dependent kinases. Here we study the phosphorylation of CDC25B at mitosis by the kinase pEg3, a member of the KIN1/PAR-1/MARK family. Using mass spectrometry analysis we demonstrate that CDC25B is phosphorylated in vitro by pEg3 on serine 169, a residue that lies within the B domain. Moreover, using phosphoepitope-specific antibodies we show that serine 169 is phosphorylated in vivo, that this phosphorylated form of CDC25B accumulates during mitosis, and is localized to the centrosomes. This labelling is abrogated when pEg3 expression is repressed by RNA interference. Taken together, these results support a model in which pEg3 contributes to the control of progression through mitosis by phosphorylation of the CDC25 phosphatases.


Assuntos
Centrossomo/metabolismo , Mitose/fisiologia , Proteínas Quinases/metabolismo , Fuso Acromático/metabolismo , Fatores de Transcrição/metabolismo , Fosfatases cdc25/metabolismo , Sequência de Aminoácidos , Células HeLa , Humanos , Fatores de Transcrição Kruppel-Like , Dados de Sequência Molecular , Fosforilação , Transporte Proteico , Proteínas Recombinantes , Serina , Análise Espectral , Fosfatases cdc25/química
20.
J Cell Sci ; 117(Pt 12): 2523-31, 2004 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15128871

RESUMO

Aurora-A protein kinase, which is the product of an oncogene, is required for the assembly of a functional mitotic apparatus and the regulation of cell ploidy. Overexpression of Aurora-A in tumour cells has been correlated with cancer susceptibility and poor prognosis. Aurora-A activity is required for the recruitment of CDK1-cyclin B1 to the centrosome prior to its activation and the commitment of the cell to mitosis. In this report, we demonstrate that the CDC25B phosphatase, an activator of cyclin dependent kinases at mitosis, is phosphorylated both in vitro and in vivo by Aurora-A on serine 353 and that this phosphorylated form of CDC25B is located at the centrosome during mitosis. Knockdown experiments by RNAi confirm that the centrosome phosphorylation of CDC25B on S353 depends on Aurora-A kinase. Microinjection of antibodies against phosphorylated S353 results in a mitotic delay whilst overexpression of a S353 phosphomimetic mutant enhances the mitotic inducing effect of CDC25B. Our results demonstrate that Aurora-A phosphorylates CDC25B in vivo at the centrosome during mitosis. This phosphorylation might locally participate in the control of the onset of mitosis. These findings re-emphasise the role of the centrosome as a functional integrator of the pathways contributing to the triggering of mitosis.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Divisão Celular/fisiologia , Centrossomo/metabolismo , Fase G2/fisiologia , Proteínas Quinases/metabolismo , Fosfatases cdc25/metabolismo , Anticorpos/metabolismo , Anticorpos Monoclonais/metabolismo , Aurora Quinases , Proteínas de Ciclo Celular/química , Células HeLa , Humanos , Microinjeções , Fosforilação , Proteínas Serina-Treonina Quinases , Interferência de RNA , Serina/metabolismo , Fatores de Tempo , Proteínas de Xenopus , Fosfatases cdc25/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...