Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Nat Hum Behav ; 7(11): 2023-2037, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37679443

RESUMEN

Despite being a topical issue in public debate and on the political agenda for many countries, a global-scale, high-resolution quantification of migration and its major drivers for the recent decades remained missing. We created a global dataset of annual net migration between 2000 and 2019 (~10 km grid, covering the areas of 216 countries or sovereign states), based on reported and downscaled subnational birth (2,555 administrative units) and death (2,067 administrative units) rates. We show that, globally, around 50% of the world's urban population lived in areas where migration accelerated urban population growth, while a third of the global population lived in provinces where rural areas experienced positive net migration. Finally, we show that, globally, socioeconomic factors are more strongly associated with migration patterns than climatic factors. While our method is dependent on census data, incurring notable uncertainties in regions where census data coverage or quality is low, we were able to capture migration patterns not only between but also within countries, as well as by socioeconomic and geophysical zonings. Our results highlight the importance of subnational analysis of migration-a necessity for policy design, international cooperation and shared responsibility for managing internal and international migration.


Asunto(s)
Emigración e Inmigración , Migración Humana , Humanos , Dinámica Poblacional , Factores Socioeconómicos , Población Urbana
3.
Clin Pharmacol Ther ; 113(2): 349-359, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36326573

RESUMEN

Important discoveries by academic drug developers hold the promise of bringing innovative treatments that address unmet medical needs to the market. However, the drug development process has proved to be challenging and demanding for academic researchers, and regulatory challenges are an important barrier to implementing academic findings in clinical practice. European regulators offer varying degrees of support services to help drug developers meet regulatory standards and requirements. "Strengthening Training of Academia in Regulatory Sciences and Supporting Regulatory Scientific Advice" (STARS) is a European Commission-funded consortium aiming to strengthen the training of academics in regulatory science and requirements. Here, we report the results of four surveys that investigated the awareness and utilization of support tools offered by European regulators and identified the regulatory challenges and support needs of researchers. The surveys targeted four main European stakeholders in academic medicines research: academic research groups (706 respondents), academic research centers (99), funding organizations (49), and regulators (22). The results show that while European regulators provide various regulatory support tools, less than half of the responding academic researchers were aware of these tools and many experienced challenges in reaching a sufficient level of regulatory knowledge. There was a general lack of understanding of the regulatory environment that was aggravated by poor communication between stakeholders. The results of this study form a foundation for an improved European medicines regulatory network, in which regulatory challenges faced by academia are tackled.


Asunto(s)
Descubrimiento de Drogas , Control de Medicamentos y Narcóticos , Humanos , Europa (Continente) , Encuestas y Cuestionarios
4.
Sci Total Environ ; 822: 153493, 2022 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-35114232

RESUMEN

Droughts are causing severe damages to tropical countries worldwide. Although water abundant, their resilience to water shortages during dry periods is often low. As there is little knowledge about tropical drought characteristics, reliable methodologies to evaluate drought risk in data scarce tropical regions are needed. We combined drought hazard and vulnerability related data to assess drought risk in four rural tropical study regions, the Muriaé basin, Southeast Brazil, the Tempisque-Bebedero basin in Costa Rica, the upper part of the Magdalena basin, Colombia and the Srepok, shared by Cambodia and Vietnam. Drought hazard was analyzed using the variables daily river discharge, precipitation and vegetation condition. Drought vulnerability was assessed based on regionally available socioeconomic data. Besides illustrating the relative severity of each indicator value, we developed drought risk maps combining hazard and vulnerability for each grid-cell. While for the Muriaé, our results identified the downstream area as being exposed to severe drought risk, the Tempisque showed highest risk along the major streams and related irrigation systems. Risk hotspots in the Upper Magdalena were found in the central valley and the dryer Southeast and in the Srepok in the agricultural areas of Vietnam and downstream Cambodia. Local scientists and stakeholders have validated our results and we believe that our drought risk assessment methodology for data scarce and rural tropical regions offers a holistic, science based and innovative framework to generate relevant drought related information. Being applied to other tropical catchments, the approaches described in this article will enable the selection of data sets, indices and their classification - depending on basin size, spatial resolution and seasonality. At its current stage, the outcomes of this study provide relevant information for regional planners and water managers dealing with the control of future drought disasters in tropical regions.


Asunto(s)
Acceso a la Información , Sequías , Agricultura , Ríos , Agua
5.
Oncoscience ; 8: 134-153, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34926718

RESUMEN

Hec1 (Highly expressed in cancer 1) resides in the outer kinetochore where it works to facilitate proper kinetochore-microtubule interactions during mitosis. Hec1 is overexpressed in various cancers and its expression shows correlation with high tumour grade and poor patient prognosis. Chemical perturbation of Hec1 is anticipated to impair kinetochore-microtubule binding, activate the spindle assembly checkpoint (spindle checkpoint) and thereby suppress cell proliferation. In this study, we performed high-throughput screen to identify novel small molecules that target the Hec1 calponin homology domain (CHD), which is needed for normal microtubule attachments. 4 million compounds were first virtually fitted against the CHD, and the best hit molecules were evaluated in vitro. These approaches led to the identification of VTT-006, a 1,2-disubstituted-tetrahydro-beta-carboline derivative, which showed binding to recombinant Ndc80 complex and modulated Hec1 association with microtubules in vitro. VTT-006 treatment resulted in chromosome congression defects, reduced chromosome oscillations and induced loss of inter-kinetochore tension. Cells remained arrested in mitosis with an active spindle checkpoint for several hours before undergoing cell death. VTT-006 suppressed the growth of several cancer cell lines and enhanced the sensitivity of HeLa cells to Taxol. Our findings propose that VTT-006 is a potential anti-mitotic compound that disrupts M phase, impairs kinetochore-microtubule interactions, and activates the spindle checkpoint.

6.
Drug Discov Today ; 26(2): 283-288, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33127567

RESUMEN

Truly disruptive medicine innovation and new treatment paradigms tend to start in non-commercial research institutions. However, the lack of mutual understanding between medicine developers and regulators when it comes to medicine development significantly delays or even prevents the access of patients to these innovations. Here, we outline what regulatory-related barriers hamper the translational development of novel products or new treatment paradigms initiated in academia, and propose key steps towards improved regulatory dialogue among academia, funding bodies and regulatory authorities. Moreover, we briefly describe how the STARS (Strengthening Training of Academia in Regulatory Science) project aims to reach out to medicine innovators in academia to bridge the regulatory knowledge gap and enhance this dialogue to facilitate the implementation of academic research findings in clinical practice.


Asunto(s)
Difusión de Innovaciones , Investigación Biomédica Traslacional/organización & administración , Tecnología Disruptiva/legislación & jurisprudencia , Unión Europea , Humanos , Investigación Biomédica Traslacional/legislación & jurisprudencia
7.
Transl Oncol ; 12(1): 170-179, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30359947

RESUMEN

Taxanes are chemotherapeutic agents used in the treatment of solid tumors, particularly of breast, ovarian, and lung origin. However, patients show divergent therapy responses, and the molecular determinants of taxane sensitivity have remained elusive. Especially the signaling pathways that promote death of the taxane-treated cells are poorly characterized. Here we describe a novel part of a signaling route in which c-Myc enhances paclitaxel sensitivity through upregulation of miR-203b-3p and miR-203a-3p; two clustered antiapoptosis protein Bcl-xL controlling microRNAs. In vitro, the miR-203b-3p decreases the expression of Bcl-xL by direct targeting of the gene's mRNA 3'UTR. Notably, overexpression of the miR-203b-3p changed the fate of paclitaxel-treated breast and ovarian cancer cells from mitotic slippage to cell death. In breast tumors, high expression of the miR-203b-3p and MYC was associated with better therapy response and patient survival. Interestingly, in the breast tumors, MYC expression correlated negatively with BCL2L1 expression but positively with miR-203b-3p and miR-203a-3p. Finally, silencing of MYC suppressed the transcription of both miRNAs in breast tumor cells. Pending further validation, these results may assist in patient stratification for taxane therapy.

8.
Drug Des Devel Ther ; 11: 1335-1351, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28496304

RESUMEN

PURPOSE: Overall, ~65% of patients diagnosed with advanced ovarian cancer (OC) will relapse after primary surgery and adjuvant first-line platinum- and taxane-based chemotherapy. Significant improvements in the treatment of OC are expected from the development of novel compounds having combined cytotoxic and antiangiogenic properties that make them effective on refractory tumors. METHODS: Permeability of NOV202 was determined with Caco-2 monolayer assay. The compound's pharmacokinetic profile and plasma:brain distribution were assessed in male C57Bl/6 mice. The compound's impacts on tubulin, microtubules and cell cycle were investigated by using in vitro tubulin polymerization assay, cell-based immunofluorescence and live cell microscopy. The IC50 concentrations of NOV202 were assessed in a panel of eight cancer cell lines. Impact of the compound on vascular tube formation was determined using the StemKit and Chick chorioallantoic membrane assays. The in vivo efficacy of the compound was analyzed with an OC xenograft mouse model. RESULTS: NOV202 was found to suppress cancer cell proliferation at low nanomolar concentrations (IC50 2.3-12.0 nM) and showed equal efficacy between OC cell line A2780 (IC50 2.4 nM) and its multidrug-resistant subline A2780/Adr (IC50 2.3 nM). Mechanistically, NOV202 targeted tubulin polymerization in vitro in a dose-dependent manner and in cells induced an M phase arrest. In vivo, NOV202 caused a dose-dependent reduction of tumor mass in an A2780 xenograft model, which at the highest dose (40 mg/kg) was comparable to the effect of paclitaxel (24 mg/kg). Interestingly, NOV202 exhibited vascular disrupting properties that were similar to the effects of Combretastatin A4. CONCLUSION: NOV202 is a novel tubulin and vascular targeting agent that shows strong anticancer efficacy in cells and OC xenograft models. The finding that the compound induced significantly more cell death in Pgp/MDR1 overexpressing OC cells compared to vincristine and paclitaxel warrants further development of the compound as a new therapy for OC patients with treatment refractory tumors and/or relapsing disease.


Asunto(s)
Antineoplásicos/farmacología , Microtúbulos/efectos de los fármacos , Neovascularización Patológica/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Neovascularización Patológica/patología , Neoplasias Ováricas/patología , Relación Estructura-Actividad , Células Tumorales Cultivadas
9.
Br J Cancer ; 116(11): 1451-1461, 2017 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-28449010

RESUMEN

BACKGROUND: Several microRNA (miRNA) molecules have emerged as important post-transcriptional regulators of tumour suppressor and oncogene expression. Ras association domain family member 1 (RASSF1) is a critical tumour suppressor that controls multiple aspects of cell proliferation such as cell cycle, cell division and apoptosis. The expression of RASSF1 is lost in a variety of cancers due to the promoter hypermethylation. METHODS: miR-193a-3p was identified as a RASSF1-targeting miRNA by a dual screening approach. In cultured human cancer cells, immunoblotting, qRT-PCR, luciferase reporter assays, time-lapse microscopy and immunofluorescence methods were used to study the effects of excess miR-193a-3p on RASSF1 expression and cell division. RESULTS: Here, we report a new miRNA-mediated mechanism that regulates RASSF1 expression: miR-193a-3p binds directly to RASSF1-3'UTR and represses the mRNA and protein expression. In human cancer cells, excess of miR-193a-3p causes polyploidy through impairment of the Rassf1-Syntaxin 16 signalling pathway that is needed for completion of cytokinesis. In the next cell cycle the miR-193a-3p-overexpressing cells exhibit multipolar mitotic spindles, mitotic delay and elevated frequency of cell death. CONCLUSIONS: Our results suggest that besides epigenetic regulation, altered expression of specific miRNAs may contribute to the loss of Rassf1 in cancer cells and cause cell division errors.


Asunto(s)
División Celular/genética , MicroARNs/genética , ARN Mensajero/metabolismo , Proteínas Supresoras de Tumor/genética , Regiones no Traducidas 3' , Muerte Celular/genética , Polaridad Celular/genética , Citocinesis/genética , Regulación hacia Abajo , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Células HeLa , Humanos , Puntos de Control de la Fase M del Ciclo Celular/genética , Transducción de Señal/genética , Sintaxina 16/metabolismo , Transfección , Proteínas Supresoras de Tumor/metabolismo
10.
FEBS Lett ; 590(16): 2757-67, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27423135

RESUMEN

Dual specificity phosphatase-3 (Dusp3/Vhr) regulates cell cycle progression by counteracting the effects of mitogen-activated protein kinases (Mapk) Erk1/2 and Jnk. Despite the known upregulation of Dusp3 at M phase in mammalian cells, its mitotic functions are poorly characterized. Here, we report that loss of Dusp3 by RNAi leads to the formation of multipolar spindles in human mitotic cancer cells in an Erk1/2-dependent manner. In the phosphatase-silenced cells, the normal bipolar spindle structure was restored by chemical inhibition of Erk1/2 and ectopic overexpression of Dusp3. We propose that at M phase Dusp3 keeps Erk1/2 activity in check to facilitate normal mitosis.


Asunto(s)
Polaridad Celular/genética , Fosfatasa 3 de Especificidad Dual/genética , Mitosis/genética , Huso Acromático/genética , Ciclo Celular/genética , Fosfatasa 3 de Especificidad Dual/biosíntesis , Regulación Neoplásica de la Expresión Génica , Células HeLa , Humanos , MAP Quinasa Quinasa 4/genética , Sistema de Señalización de MAP Quinasas/genética , Fosforilación , Interferencia de ARN , Transfección
11.
Oncotarget ; 7(11): 12267-85, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26943585

RESUMEN

The molecular pathways that contribute to the proliferation and drug response of cancer cells are highly complex and currently insufficiently characterized. We have identified a previously unknown microRNA-based mechanism that provides cancer cells means to stimulate tumorigenesis via increased genomic instability and, at the same time, evade the action of clinically utilized microtubule drugs. We demonstrate miR-493-3p to be a novel negative regulator of mitotic arrest deficient-2 (MAD2), an essential component of the spindle assembly checkpoint that monitors the fidelity of chromosome segregation. The microRNA targets the 3' UTR of Mad2 mRNA thereby preventing translation of the Mad2 protein. In cancer cells, overexpression of miR-493-3p induced a premature mitotic exit that led to increased frequency of aneuploidy and cellular senescence in the progeny cells. Importantly, excess of the miR-493-3p conferred resistance of cancer cells to microtubule drugs. In human neoplasms, miR-493-3p and Mad2 expression alterations correlated with advanced ovarian cancer forms and high miR-493-3p levels were associated with reduced survival of ovarian and breast cancer patients with aggressive tumors, especially in the paclitaxel therapy arm. Our results suggest that intratumoral profiling of miR-493-3p and Mad2 levels can have diagnostic value in predicting the efficacy of taxane chemotherapy.


Asunto(s)
Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Proteínas Mad2/metabolismo , MicroARNs/metabolismo , Neoplasias/tratamiento farmacológico , Paclitaxel/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Segregación Cromosómica , Femenino , Células HCT116 , Células HeLa , Humanos , Proteínas Mad2/genética , MicroARNs/genética , Mitosis/efectos de los fármacos , Mitosis/fisiología , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Ensayos Clínicos Controlados Aleatorios como Asunto , Transfección
12.
Mol Oncol ; 9(6): 1056-70, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25682900

RESUMEN

The let-7 microRNA (miRNA) family has been implicated in the regulation of diverse cellular processes and disease pathogenesis. In cancer, loss-of-function of let-7 miRNAs has been linked to tumorigenesis via increased expression of target oncogenes. Excessive proliferation rate of tumor cells is often associated with deregulation of mitotic proteins. Here, we show that let-7b contributes to the maintenance of genomic balance via targeting Aurora B kinase, a key regulator of the spindle assembly checkpoint (SAC). Our results indicate that let-7b binds to Aurora B kinase 3'UTR reducing mRNA and protein expression of the kinase. In cells, excess let-7b induced mitotic defects characteristic to Aurora B perturbation including increased rate of polyploidy and multipolarity, and premature SAC inactivation that leads to forced exit from chemically induced mitotic arrest. Moreover, the frequency of aneuploid HCT-116 cells was significantly increased upon let-7b overexpression compared to controls. Interestingly, together with a chemical Aurora B inhibitor, let-7b had an additive effect on polyploidy induction in HeLa cells. In breast cancer patients, reduced let-7b expression was found to be associated with increased Aurora B expression in grade 3 tumors. Furthermore, let-7b was found downregulated in the most aggressive forms of breast cancer determined by clinicopathological parameters. Together, our findings suggest that let-7b contributes to the fidelity of cell division via regulation of Aurora B. Moreover, the loss of let-7b in aggressive tumors may drive tumorigenesis by up-regulation of Aurora B and other targets of the miRNA, which further supports the role of let-7b in tumor suppression.


Asunto(s)
Aurora Quinasa B/metabolismo , Neoplasias de la Mama/metabolismo , MicroARNs/metabolismo , Proteínas de Neoplasias/metabolismo , Poliploidía , ARN Neoplásico/metabolismo , Regiones no Traducidas 3' , Aurora Quinasa B/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Femenino , Células HeLa , Humanos , MicroARNs/genética , Proteínas de Neoplasias/genética , ARN Neoplásico/genética
13.
J Cell Biol ; 206(6): 735-49, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25202032

RESUMEN

Unless mitigated, external and physiological stresses are detrimental for cells, especially in mitosis, resulting in chromosomal missegregation, aneuploidy, or apoptosis. Heat shock proteins (Hsps) maintain protein homeostasis and promote cell survival. Hsps are transcriptionally regulated by heat shock factors (HSFs). Of these, HSF1 is the master regulator and HSF2 modulates Hsp expression by interacting with HSF1. Due to global inhibition of transcription in mitosis, including HSF1-mediated expression of Hsps, mitotic cells are highly vulnerable to stress. Here, we show that cells can counteract transcriptional silencing and protect themselves against proteotoxicity in mitosis. We found that the condensed chromatin of HSF2-deficient cells is accessible for HSF1 and RNA polymerase II, allowing stress-inducible Hsp expression. Consequently, HSF2-deficient cells exposed to acute stress display diminished mitotic errors and have a survival advantage. We also show that HSF2 expression declines during mitosis in several but not all human cell lines, which corresponds to the Hsp70 induction and protection against stress-induced mitotic abnormalities and apoptosis.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Choque Térmico/genética , Respuesta al Choque Térmico/genética , Mitosis/genética , ARN Polimerasa II/genética , Factores de Transcripción/genética , Animales , Apoptosis/genética , Línea Celular Tumoral , Supervivencia Celular , Cromatina/genética , Regulación de la Expresión Génica , Proteínas HSP70 de Choque Térmico/biosíntesis , Proteínas HSP70 de Choque Térmico/genética , Células HeLa , Factores de Transcripción del Choque Térmico , Proteínas de Choque Térmico/biosíntesis , Humanos , Células MCF-7 , Ratones , Índice Mitótico , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Interferente Pequeño , Factores de Transcripción/biosíntesis , Transcripción Genética
14.
PLoS One ; 9(8): e105526, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25148385

RESUMEN

Although c-Abl has increasingly emerged as a key player in the DNA damage response, its role in this context is far from clear. We studied the effect of inhibition of c-Abl kinase activity by imatinib with chemotherapy drugs and found a striking difference in cell survival after combined mitoxantrone (MX) and imatinib treatment compared to a panel of other chemotherapy drugs. The combinatory treatment induced apoptosis in HeLa cells and other cancer cell lines but not in primary fibroblasts. The difference in MX and doxorubicin was related to significant augmentation of DNA damage. Transcriptionally active p53 accumulated in cells in which human papillomavirus E6 normally degrades p53. The combination treatment resulted in caspase activation and apoptosis, but this effect did not depend on either p53 or p73 activity. Despite increased p53 activity, the cells arrested in G2 phase became defective in this checkpoint, allowing cell cycle progression. The effect after MX treatment depended partially on c-Abl: Short interfering RNA knockdown of c-Abl rendered HeLa cells less sensitive to MX. The effect of imatinib was decreased by c-Abl siRNA suggesting a role for catalytically inactive c-Abl in the death cascade. These findings indicate that MX has a unique cytotoxic effect when the kinase activity of c-Abl is inhibited. The treatment results in increased DNA damage and c-Abl-dependent apoptosis, which may offer new possibilities for potentiation of cancer chemotherapy.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Mitoxantrona/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-abl/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Benzamidas/farmacología , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Línea Celular Tumoral , Daño del ADN/efectos de los fármacos , Regulación hacia Abajo , Resistencia a Antineoplásicos/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Mesilato de Imatinib , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-abl/genética , Proteínas Proto-Oncogénicas c-abl/metabolismo , Pirimidinas/farmacología , Interferencia de ARN , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos , Inhibidores de Topoisomerasa II/farmacología , Proteína p53 Supresora de Tumor/metabolismo
15.
Mol Cancer Ther ; 13(5): 1054-66, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24748653

RESUMEN

Mitosis is an attractive target for the development of new anticancer drugs. In a search for novel mitotic inhibitors, we virtually screened for low molecular weight compounds that would possess similar steric and electrostatic features, but different chemical structure than rigosertib (ON 01910.Na), a putative inhibitor of phosphoinositide 3-kinase (PI3K) and polo-like kinase 1 (Plk1) pathways. Highest scoring hit compounds were tested in cell-based assays for their ability to induce mitotic arrest. We identified a novel acridinyl-acetohydrazide, here named as Centmitor-1 (Cent-1), that possesses highly similar molecular interaction field as rigosertib. In cells, Cent-1 phenocopied the cellular effects of rigosertib and caused mitotic arrest characterized by chromosome alignment defects, multipolar spindles, centrosome fragmentation, and activated spindle assembly checkpoint. We compared the effects of Cent-1 and rigosertib on microtubules and found that both compounds modulated microtubule plus-ends and reduced microtubule dynamics. Also, mitotic spindle forces were affected by the compounds as tension across sister kinetochores was reduced in mitotic cells. Our results showed that both Cent-1 and rigosertib target processes that occur during mitosis as they had immediate antimitotic effects when added to cells during mitosis. Analysis of Plk1 activity in cells using a Förster resonance energy transfer (FRET)-based assay indicated that neither compound affected the activity of the kinase. Taken together, these findings suggest that Cent-1 and rigosertib elicit their antimitotic effects by targeting mitotic processes without impairment of Plk1 kinase activity.


Asunto(s)
Acridonas/farmacología , Antimitóticos/farmacología , Glicina/análogos & derivados , Hidrazinas/farmacología , Sulfonas/farmacología , Acridonas/química , Antimitóticos/química , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/antagonistas & inhibidores , Centrosoma/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Glicina/química , Glicina/farmacología , Células HeLa , Ensayos Analíticos de Alto Rendimiento , Humanos , Hidrazinas/química , Microtúbulos/metabolismo , Mitosis/efectos de los fármacos , Estructura Molecular , Peso Molecular , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Sulfonas/química , Quinasa Tipo Polo 1
17.
Chromosoma ; 122(5): 431-49, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23775312

RESUMEN

Suppression of cell proliferation by targeting mitosis is one potential cancer intervention. A number of existing chemotherapy drugs disrupt mitosis by targeting microtubule dynamics. While efficacious, these drugs have limitations, i.e. neuropathy, unpredictability and development of resistance. In order to overcome these issues, a great deal of effort has been spent exploring novel mitotic targets including Polo-like kinase 1, Aurora kinases, Mps1, Cenp-E and KSP/Eg5. Here we summarize the latest developments in the discovery and clinical evaluation of new mitotic drug targets.


Asunto(s)
Anticarcinógenos/uso terapéutico , Mitosis/genética , Terapia Molecular Dirigida , Aurora Quinasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Proteínas Cromosómicas no Histona/metabolismo , Humanos , Cinesinas/metabolismo , Mitosis/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Quinasa Tipo Polo 1
18.
Carcinogenesis ; 34(2): 436-45, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23104179

RESUMEN

Mitosis represents a clinically important determination point in the life cycle of proliferating cells. One potential drug target within the mitotic machinery is the spindle assembly checkpoint (SAC), an evolutionarily conserved signaling pathway that monitors the connections between microtubules (MTs) and chromosomes. Mistakes in SAC signaling may lead to cell division errors that can trigger elimination of cancer cells at M phase or soon after exit from mitosis. In this study, we describe the cellular effects of a novel pyrimidine-2,4-diamine derivative that we discovered to inhibit the activity of SAC. The compound caused rapid escape from the mitotic arrest induced by lack of interkinetochore tension but not by lack of MT-kinetochore attachments. In cycling cells, the compound disrupted the architecture of mitotic spindle that triggered a transient M-phase arrest that was rapidly followed by a forced mitotic exit. The premature termination of M phase was found to be a consequence of precocious inactivation of SAC caused by a direct inhibitory effect of the compound on Aurora B kinase in vitro and in cells. The compound also targets Aurora A kinase and tubulin in vitro and in cells, which can explain the observed spindle anomalies. The reduced activity of Aurora B kinase resulted in polyploidy and suppression of cancer cell viability. Our data suggest that this new pharmacophore possesses interesting anticancer properties that could be exploited in development of mitosis-targeting therapies.


Asunto(s)
Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Neoplasias/patología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pirimidinas/farmacología , Huso Acromático/efectos de los fármacos , Aurora Quinasa B , Aurora Quinasas , Western Blotting , Técnica del Anticuerpo Fluorescente , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Células Tumorales Cultivadas , Ensayo de Tumor de Célula Madre
19.
Exp Cell Res ; 318(5): 578-92, 2012 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-22227008

RESUMEN

The spindle assembly checkpoint (SAC) is a conserved mechanism that ensures the fidelity of chromosome distribution in mitosis by preventing anaphase onset until the correct bipolar microtubule-kinetochore attachments are formed. Errors in SAC function may contribute to tumorigenesis by inducing numerical chromosome anomalies (aneuploidy). On the other hand, total disruption of SAC can lead to massive genomic imbalance followed by cell death, a phenomena that has therapeutic potency. We performed a cell-based high-throughput screen with a compound library of 2000 bioactives for novel SAC inhibitors and discovered a plant-derived phenolic compound eupatorin (3',5-dihydroxy-4',6,7-trimethoxyflavone) as an anti-mitotic flavonoid. The premature override of the microtubule drug-imposed mitotic arrest by eupatorin is dependent on microtubule-kinetochore attachments but not interkinetochore tension. Aurora B kinase activity, which is essential for maintenance of normal SAC signaling, is diminished by eupatorin in cells and in vitro providing a mechanistic explanation for the observed forced mitotic exit. Eupatorin likely has additional targets since eupatorin treatment of pre-mitotic cells causes spindle anomalies triggering a transient M phase delay followed by impaired cytokinesis and polyploidy. Finally, eupatorin potently induces apoptosis in multiple cancer cell lines and suppresses cancer cell proliferation in organotypic 3D cell culture model.


Asunto(s)
Antimitóticos/farmacología , Apoptosis/efectos de los fármacos , Flavonoides/farmacología , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Poliploidía , Aurora Quinasa B , Aurora Quinasas , Técnicas de Cultivo de Célula , Proliferación Celular/efectos de los fármacos , Centrosoma/metabolismo , Células HeLa , Ensayos Analíticos de Alto Rendimiento , Humanos , Leupeptinas/farmacología , Masculino , Microscopía Fluorescente , Nocodazol/farmacología , Neoplasias de la Próstata , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Pirimidinas/farmacología , Tionas/farmacología , Imagen de Lapso de Tiempo
20.
Mol Oncol ; 5(1): 105-11, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21051298

RESUMEN

Cell division is orchestrated by a complex protein network that aims to maintenance of genomic stability. Visualisation of mitotic protein-protein associations in space and time has been limited due to the lack of proper biochemical and easy-to-use imaging tools. Here we report adaptation of the in situ proximity ligation assay (is-PLA) to study mitotic protein interactions with spatio-temporal resolution. We examined the composition of the Chromosomal Passenger Complex (CPC) at various mitotic phases and after chemical treatments using is-PLA with antibodies against the core CPC subunits Aurora B, INCENP, Survivin and Borealin. Our results support the notion that the core CPC functions as a single structural unit at centromeres in early mitosis and at central spindle after the onset of anaphase. Treatment of cells with the Aurora B inhibitor ZM447439 diminished the is-PLA signals at centromeres suggesting that Aurora B activity contributes to structural maintenance and/or proper subcellular localization of the core CPC. Is-PLA-based analysis of interaction between INCENP and Polo-like kinase 1 (Plk1) proposes that the kinase co-travels with CPC during late mitosis. The data illustrates both the strengths and limitations of the is-PLA in the analysis of mitotic macromolecule associations at sub-organelle level.


Asunto(s)
Cromosomas Humanos , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA