Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 8(1): 607, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28928360

RESUMEN

In melanoma, therapies with inhibitors to oncogenic BRAFV600E are highly effective but responses are often short-lived due to the emergence of drug-resistant tumor subpopulations. We describe here a mechanism of acquired drug resistance through the tumor microenvironment, which is mediated by human tumor-associated B cells. Human melanoma cells constitutively produce the growth factor FGF-2, which activates tumor-infiltrating B cells to produce the growth factor IGF-1. B-cell-derived IGF-1 is critical for resistance of melanomas to BRAF and MEK inhibitors due to emergence of heterogeneous subpopulations and activation of FGFR-3. Consistently, resistance of melanomas to BRAF and/or MEK inhibitors is associated with increased CD20 and IGF-1 transcript levels in tumors and IGF-1 expression in tumor-associated B cells. Furthermore, first clinical data from a pilot trial in therapy-resistant metastatic melanoma patients show anti-tumor activity through B-cell depletion by anti-CD20 antibody. Our findings establish a mechanism of acquired therapy resistance through tumor-associated B cells with important clinical implications.Resistance to BRAFV600E inhibitors often occurs in melanoma patients. Here, the authors describe a potential mechanism of acquired drug resistance mediated by tumor-associated B cells-derived IGF-1.


Asunto(s)
Antineoplásicos/uso terapéutico , Linfocitos B/metabolismo , Resistencia a Antineoplásicos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Linfocitos Infiltrantes de Tumor/metabolismo , Melanoma/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Neoplasias Cutáneas/tratamiento farmacológico , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Supervivencia Celular , Cisplatino/uso terapéutico , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Humanos , Técnicas In Vitro , Melanoma/genética , Paclitaxel/uso terapéutico , Proyectos Piloto , Proteínas Proto-Oncogénicas B-raf/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Neoplasias Cutáneas/genética , Microambiente Tumoral
2.
Mutagenesis ; 31(5): 573-82, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27180333

RESUMEN

Current therapy approaches in melanoma targeting have met with the development of resistance and tumour recurrence with a more aggressive phenotype. In a quest for alternative therapy targets, we had previously identified Signal Sequence Receptor 2 (SSR2) as a gene with high expression in a subgroup of human primary melanomas. Now we show that SSR2 exerts a prosurvival functionality in human melanoma cells and that high expression levels of SSR2 are associated with an unfavourable disease outcome in primary melanoma patients. Consistent with SSR's role in translocation of proteins from the ribosome across the endoplasmic reticulum (ER) membrane, our data supports induction of SSR2 as a part of the ER stress response. This response included SSR2 upregulation upon development of therapy resistance to BRAF inhibitors, as well as the dependency of cell survival of BRAF inhibitor-resistant melanoma cells on SSR2. Complementary gain and loss of function data showed the Unfolded Protein Response (UPR) to ER stress as an inducer of SSR2 via transcriptional regulation through X-Box Binding Protein 1s (XBP1s) and support an ER stress-UPR-Transcription Factor XBP1s-SSR2 response axis in human melanocytic cells. Together with its dispensability for survival in normal human cells, these data propose SSR2 as a potential therapeutic target in (therapy-resistant) human melanoma.


Asunto(s)
Proteínas de Unión al Calcio/genética , Estrés del Retículo Endoplásmico , Melanoma/metabolismo , Glicoproteínas de Membrana/genética , Receptores Citoplasmáticos y Nucleares/genética , Receptores de Péptidos/genética , Activación Transcripcional , Respuesta de Proteína Desplegada , Línea Celular , Supervivencia Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/fisiopatología
3.
Hum Pathol ; 54: 157-64, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27107457

RESUMEN

B cells often infiltrate the microenvironment of human tumors. B cells can both positively and negatively regulate antitumor immune responses. In several human cancers, higher numbers of CD20(+) TAB are associated with a favorable prognosis, whereas in human primary melanomas, this association is contentious. In this study, we determined the association of TAB numbers in cutaneous primary melanoma tissue samples and patients' overall survival. The CD20 immunohistochemistry on archival nonmetastasized and metastasized cutaneous primary melanoma tissues from 2 independent patient cohorts was performed. One cohort was used in class comparison for metastasis, the most important prognostic factor for overall survival, and the other cohort for a subsequent survival analysis. Survival association was further validated with RNA data from a third independent cohort. Whole tissue sections were read automatically via quantitative digital imaging and analysis. Survival data were analyzed by Cox proportional hazard modeling. We discovered that cutaneous primary melanomas without metastasis contain significantly more TAB than primary melanomas that had metastasized. At time of first diagnosis, a higher number of TAB is associated with a significantly better overall survival in patients with cutaneous primary melanomas of >1 mm Breslow depth. Also, higher CD20/CD19 tumor mRNA levels are correlated with a significantly better overall survival. Thus, our data support TAB numbers as a prognostic biomarker in cutaneous primary melanoma patients with a tumor of >1 mm Breslow depth. For a survey in larger studies, whole tissue section analysis seems to be key to accurate assessment of TAB numbers.


Asunto(s)
Linfocitos B/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma/inmunología , Neoplasias Cutáneas/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD19/análisis , Antígenos CD19/genética , Antígenos CD20/análisis , Antígenos CD20/genética , Austria , Linfocitos B/patología , Biomarcadores de Tumor/análisis , Bases de Datos Genéticas , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Recuento de Linfocitos , Linfocitos Infiltrantes de Tumor/patología , Masculino , Melanoma/genética , Melanoma/mortalidad , Melanoma/secundario , Persona de Mediana Edad , Invasividad Neoplásica , Valor Predictivo de las Pruebas , Pronóstico , Modelos de Riesgos Proporcionales , Reproducibilidad de los Resultados , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/patología , Suiza , Factores de Tiempo , Microambiente Tumoral
4.
J Proteome Res ; 13(6): 2830-45, 2014 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-24803318

RESUMEN

Melanoma, the deadliest form of skin cancer, is highly immunogenic and frequently infiltrated with immune cells including B cells. The role of tumor-infiltrating B cells (TIBCs) in melanoma is as yet unresolved, possibly due to technical challenges in obtaining TIBCs in sufficient quantity for extensive studies and due to the limited life span of B cells in vitro. A comprehensive workflow has thus been developed for successful isolation and proteomic analysis of a low number of TIBCs from fresh, human melanoma tissue. In addition, we generated in vitro-proliferating TIBC cultures using simultaneous stimulation with Epstein-Barr virus (EBV) and the TLR9 ligand CpG-oligodesoxynucleotide (CpG ODN). The FASP method and iTRAQ labeling were utilized to obtain a comparative, semiquantitative proteome to assess EBV-induced changes in TIBCs. By using as few as 100 000 B cells (∼5 µg protein)/sample for our proteomic study, a total number of 6507 proteins were identified. EBV-induced changes in TIBCs are similar to those already reported for peripheral B cells and largely involve changes in cell cycle proliferation, apoptosis, and interferon response, while most of the proteins were not significantly altered. This study provides an essential, further step toward detailed characterization of TIBCs including functional in vitro analysis.


Asunto(s)
Linfocitos B/metabolismo , Herpesvirus Humano 4/fisiología , Melanoma/inmunología , Proteoma/metabolismo , Linfocitos B/virología , Proliferación Celular , Separación Celular , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Ontología de Genes , Humanos , Melanoma/patología , Melanoma/secundario , Anotación de Secuencia Molecular , Proteoma/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Células Tumorales Cultivadas
5.
J Proteome Res ; 12(2): 1040-8, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23214492

RESUMEN

The performance of two proteomic sample preparation methods, "pseudoshotgun" (PSG) and filter-aided sample preparation (FASP) were compared in terms of the number of identified proteins, representation of cellular component GO (gene ontology) categories in the obtained list of proteins, and the efficiency of both methods in the proteomic analysis of a very low number of cells. Both methods were combined to obtain a proteomic profile of a short-term culture (passage 3) of melanoma cells, established in our laboratory from a human metastatic melanoma lesion. The data revealed that with FASP, usually more proteins are identified than with PSG when analyzing a higher number of cells (≥ 5000/injection), whereas PSG is favorable when analyzing only a very small amount of cells (250-500/injection). PSG and FASP, however, are complementary techniques, as combining both methods further increases the number of identified proteins. Moreover, we show that it is feasible to identify a substantial number of proteins from only 250 cells/injection that is equivalent to 60 ng of protein.


Asunto(s)
Melanoma/química , Proteínas de Neoplasias/aislamiento & purificación , Fragmentos de Péptidos/aislamiento & purificación , Proteoma/aislamiento & purificación , Neoplasias Cutáneas/química , Recuento de Células , Cromatografía Liquida , Filtración/métodos , Humanos , Límite de Detección , Melanoma/secundario , Proteínas de Neoplasias/química , Proteolisis , Proteoma/química , Neoplasias Cutáneas/patología , Espectrometría de Masas en Tándem , Tripsina/química , Células Tumorales Cultivadas
6.
Oncoimmunology ; 1(8): 1454-1456, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23243627

RESUMEN

Several melanoma cell subpopulations with tumor-initiating and/or tumor-maintaining properties exist that may contribute to chemoresistance and tumor recurrence after standard therapies. One of these subpopulations expresses a B-cell marker, CD20. In a small pilot trial, we showed that a subset of Stage IV melanoma patients may potentially benefit from an adjuvant treatment using the anti-CD20 antibody rituximab.

7.
Biochem Pharmacol ; 84(10): 1318-31, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22906755

RESUMEN

Two cellular proteins encoded by the breast and ovarian cancer type 1 susceptibility (BRCA1 and BRCA2) tumor suppressor genes are essential for DNA integrity and the maintenance of genomic stability. Approximately 5-10% of breast and ovarian cancers result from inherited alterations or mutations in these genes. Remarkably, BRCA1/BRCA2-deficient cells are hypersensitive to selective inhibition of poly(ADP-ribose)polymerase 1 (PARP-1), whose primary functions are related to DNA base excision repair; PARP-1 inhibition significantly potentiates the cytotoxicity of various anti-cancer drugs, including inhibitors of topoisomerase I and II. In the present study, we examined the anti-proliferative and pro-apoptotic effects of C-1305, a selective inhibitor of topoisomerase II, on human breast cancer cell lines with different BRCA1 and p53 statuses. BRCA1-competent breast cancer cell lines exhibited different responses to topoisomerase II inhibition. BT-20 cells that express high levels of BRCA1 levels were most resistant to C-1305 than other tested cells. Surprisingly, pharmacological interference with PARP-1 activity strongly inhibited their proliferation and potentiated the efficacy of C-1305 treatment. In contrast, PARP-1 inhibition only weakly affected the proliferation of BRCA1-deficient SKBr-3 cells and was not synergistic with the effects of C-1305. Further experiments revealed that the inhibition of PARP-1 in BT-20 cells caused the accumulation of DNA strand breaks and induced caspase-3 dependent apoptosis. These results seem to indicate that PARP-1 inhibition can potentiate the cytotoxicity of anti-cancer drugs in cancer cells with functional BRCA1 and suggest that mutations in other DNA repair proteins may render cancer cells more sensitive to interference with PARP-1 activity.


Asunto(s)
Acridinas/farmacología , Antineoplásicos/farmacología , Proteína BRCA1/metabolismo , Inhibidores de Topoisomerasa II/farmacología , Triazoles/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama , Caspasa 3/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Roturas del ADN , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Femenino , Humanos , Poli(ADP-Ribosa) Polimerasa-1 , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
8.
J Exp Ther Oncol ; 9(1): 17-25, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21275262

RESUMEN

Roscovitine (ROSC), a selective inhibitor of cyclin-dependent kinases (CDKs) reduces numbers of cancer cells in a concentration-dependent manner. At low doses ROSC arrests cell cycle progression and at higher doses it induces apoptosis. ROSC efficiently inhibits proliferation of human ER-alpha positive MCF-7 breast cancer cells by inducing G/M arrest and concomitantly initiates apoptosis by a p53-dependent pathway. However, the effect of ROSC is much weaker on MCF-7 cells maintained in the presence of estrogen-mimicking compounds. Therefore, we have examined the action of ROSC on other breast cancer cell lines differing in ER status and confirmed that tamoxifen (TAM) affects the efficacy of this CDK inhibitor. ROSC was effective against all tested breast cancer cell lines, arresting them at G1/S or G2/M transition and inducing apoptosis in SKBR-3 cells. Interestingly, TAM affected all tested cell lines, irrespective of their ER-a status, and in combination with ROSC it enhanced G1 or G2 arrest. Our results provide evidence that ROSC can be combined with antiestrogen therapy and that the mode of ROSC action strongly depends on the cellular context. The effect of TAM on ER-negative cancer cells indicates that TAM also crosstalks with other steroid hormone receptors.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Receptor alfa de Estrógeno/metabolismo , Fase G1/efectos de los fármacos , Fase G2/efectos de los fármacos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Femenino , Citometría de Flujo , Humanos , Immunoblotting , Purinas/administración & dosificación , Roscovitina , Tamoxifeno/administración & dosificación , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/metabolismo
9.
J Exp Ther Oncol ; 9(1): 5-15, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21275261

RESUMEN

Triazoloacridone C-1305, a new topoisomerase II inhibitor, exhibits potent cytostatic activity toward various tumours under in vitro and in vivo conditions. Interestingly, mouse cells lacking poly(ADP-ribose) polymerase-1 are much more sensitive to C-1305 than their normal counterparts. In the present study we tested the hypothesis that the functional status of p53 in tumour cells might have an impact on the efficiency of C-1305 in experiments with both p53-deficient human HL-60 promyelocytic leukemia cells and human MCF-7 breast cancer cells harboring a functional p53 pathway. Exposure of both cancer cell lines to C-1305 reduced the number of viable cells in a time- and concentration-dependent manner. Remarkably, however, HL-60 cells were much more strongly affected than MCF-7 cells. Measurements of DNA concentrations in single cells revealed that C-1305 arrested the tested cancer cells at the G/M transition. Analysis of the cell cycle and apoptosis regulators revealed that C-1305 strongly elevated phosphorylation of CDK1 at the inhibitory sites (Thr14/Tyr15) in HL-60 cells. Furthermore, C-1305 increased phosphorylation of pRb protein and CDK2 at Thr160 in HL60 cells, but not in MCF-7 cells. These observations suggest that C-1305 abrogates the restriction checkpoint and promotes G1/S transition in cells lacking functional p53.


Asunto(s)
Acridinas/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Fase G2/efectos de los fármacos , Inhibidores de Topoisomerasa II/farmacología , Triazoles/farmacología , Proteína p53 Supresora de Tumor/fisiología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Quinasa 2 Dependiente de la Ciclina/metabolismo , Femenino , Células HL-60/patología , Humanos , Immunoblotting , Fosforilación/efectos de los fármacos , Células Tumorales Cultivadas
10.
J Exp Ther Oncol ; 9(1): 27-35, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21275263

RESUMEN

Small molecule inhibitors of cyclin-dependent kinases (CDKs) show high therapeutic potential against rapidly dividing cancers and malignancies, characterized by the accumulation of transformed cells due to deregulation of apoptosis, such as multiple myeloma. In the present study we addressed the possibility that pharmacological CDK inhibitors like Roscovitine (ROSC) may be effective against human multiple myeloma cells that have acquired resistance to doxorubicin (DOX). For this purpose we selected an experimental model of human multiple myeloma-sensitive (RPMI-8226s) and doxorubicin-resistant (RPMI-8226(DOX40)) cell lines. Exposure of RPMI-8826 cells to ROSC markedly increased the proportion of hypoploid cells, representing cells undergoing apoptosis, in both sensitive and resistant cells. Unlike ROSC, DOX at high dosage did not elevate the apoptosis rate in the RPMI-8226(DOX40) cell line. Our results show that ROSC has the capacity to induce apoptosis in the RPMI-8226(DOX40) cells overexpressing the P-gp glycoprotein. Since ROSC not only inhibits cell cycle-related CDKs but also negatively regulates kinases involved in the regulation of transcription, its administration to quiescent multidrug-resistant cells might be advantageous. Inhibition of transcription of pro-survival genes such as BCL2 and MCL-1 as well as destabilization of survivin seems to improve its therapeutic efficacy.


Asunto(s)
Apoptosis/efectos de los fármacos , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Resistencia a Múltiples Medicamentos , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/patología , Inhibidores de Proteínas Quinasas/uso terapéutico , Purinas/uso terapéutico , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Antibióticos Antineoplásicos/farmacología , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Citometría de Flujo , Humanos , Immunoblotting , Mieloma Múltiple/metabolismo , Roscovitina , Células Tumorales Cultivadas
11.
J Exp Ther Oncol ; 9(1): 37-45, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21275264

RESUMEN

We reported recently that roscovitine (ROSC), a selective cyclin-dependent kinase (CDK) inhibitor, can arrest human ER-positive MCF-7 breast cancer cells in the G2 phase of the cell cycle and concomitantly induce apoptosis. The observed effects of ROSC were diminished in MCF-7 cells maintained in the presence of estrogen-mimicking compounds. Therefore, we decided to test whether combining ROSC with anti-estrogen therapy would modulate the efficacy of ROSC action. Exposure of MCF-7 cells to tamoxifen (TAM) for 24 h decreased the number of living cells by approximately 10%. This was associated with a ca. 25% increase in the G1 cell population and reduction in the proportion of S-phase cells. Unlike TAM, estrogen had very weak effects on the cell cycle progression of MCF-7 cells within 24 h. The proliferation-promoting effect of estrogen did not become evident until cultivation of cells for 48 h. Addition of estrogen to MCF-7 cells 1 h prior to TAM administration abolished the anti-estrogen-induced G1 arrest. Simultaneous treatment of MCF-7 cells with ROSC and TAM strongly enhanced the anti-proliferative effect of ROSC. This was potentiated after co-treatment with estrogen. These results clearly indicate that the efficacy of treating ER-positive breast cancers by ROSC can be enhanced by combined application of antiestrogens.


Asunto(s)
Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Receptor alfa de Estrógeno/metabolismo , Fase G1/efectos de los fármacos , Mieloma Múltiple/tratamiento farmacológico , Fase S/efectos de los fármacos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Sinergismo Farmacológico , Citometría de Flujo , Humanos , Immunoblotting , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Purinas/administración & dosificación , Roscovitina , Tamoxifeno/administración & dosificación , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/metabolismo
12.
J Cell Biochem ; 112(3): 761-72, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21328450

RESUMEN

Roscovitine (ROSC), a selective cyclin-dependent kinase (CDK) inhibitor, arrests human estrogen receptor-α (ER-α) positive MCF-7 breast cancer cells in the G(2) phase of the cell cycle and concomitantly induces apoptosis via a p53-dependent pathway. The effect of ROSC is markedly diminished in MCF-7 cells maintained in the presence of estrogen-mimicking compounds. Therefore, we decided to examine whether ROSC has any effect on the functional status of the ER-α transcription factor. Exposure of MCF-7 cells to ROSC abolished the activating phosphorylation of CDK2 and CDK7 in a concentration and time-dependent manner. This inhibition of site-specific modification of CDK7 at Ser164/170 prevented phosphorylation of RNA polymerase II and reduced basal phosphorylation of ER-α at Ser118 in non-stimulated MCF-7 cells (resulting in its down-regulation). In MCF-7 cells, estrogen induced strong phosphorylation of ER-α at Ser118 but not at Ser104/Ser106. ROSC prevented this estrogen-promoted activating modification of ER-α. Furthermore, we sought to determine whether the activity of ROSC could be enhanced by combining it with an anti-estrogen. Tamoxifen (TAM), a selective estrogen receptor modulator (SERM), affected breast cancer cell lines irrespective of their ER status. In combination with ROSC, however, it had a different impact, enhancing G(1) or G(2) arrest. Our results indicate that ROSC prevents the activating phosphorylation of ER-α and that its mode of action is strongly dependent on the cellular context. Furthermore, our data show that ROSC can be combined with anti-estrogen therapy. The inhibitory effect of TAM on ER-negative cancer cells indicates that SERMs crosstalk with other steroid hormone receptors.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/metabolismo , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Receptor alfa de Estrógeno/metabolismo , Neoplasias Hormono-Dependientes/metabolismo , Purinas/farmacología , Antineoplásicos Hormonales/farmacología , Neoplasias de la Mama/patología , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Diclororribofuranosil Benzoimidazol/farmacología , Sinergismo Farmacológico , Inhibidores Enzimáticos/farmacología , Estradiol/farmacología , Estrógenos/farmacología , Femenino , Humanos , Neoplasias Hormono-Dependientes/patología , Fosforilación , ARN Polimerasa II/metabolismo , Roscovitina , Tamoxifeno/farmacología , Proteína p53 Supresora de Tumor/metabolismo
13.
Curr Pharm Des ; 17(3): 256-71, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21348827

RESUMEN

The deregulation of apoptosis and the cell cycle are important steps in the onset of cancer, giving cells unlimited reproductive potential and increasing their likelihood of survival. The cell cycle is an essential and tightly regulated four-stage process that effects the accurate duplication and transmission of genetic content to cells' progeny. Cyclin-dependent kinases (CDKs) are key elements of the mammalian cell cycle machinery. Their activity is normally regulated via cyclin binding, phosphorylation events, and interactions with endogenous CDK inhibitors. Malfunctions in the control of the cell cycle can be specifically countered using pharmacological CDK inhibitors. Importantly, CDK inhibitors are very effective against both rapidly dividing and quiescent cancer cells; this is particularly relevant in the treatment of malignancies such as chronic lymphatic leukemia (CLL) and multiple myeloma (MM) that exhibit both a low mitotic index and apoptotic defects. The high efficacy of pharmacological CDK inhibitors against CLL and MM is attributable to their ability to eliminate leukemic cells by apoptosis. Indeed, not only do pharmacological CDK inhibitors block cell cycle progression; they also promote apoptosis and thereby destroy irrevocably malignant cells. This article focuses on the impact of inhibiting individual cellular CDKs on apoptosis. We discuss in detail the molecular mechanisms by which CDK inhibitors are able to bypass chemoresistance in tumor cells and trigger apoptosis. Remarkably, recent findings suggest that the pharmacological utility of CDK inhibitors may not be restricted to the treatment of cancer: some may be efficacious in the treatment of patients with neurodegenerative and cardiovascular diseases.


Asunto(s)
Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Animales , Quinasas Ciclina-Dependientes/metabolismo , Humanos , Ratones , Terapia Molecular Dirigida , Neoplasias/patología , Neoplasias/fisiopatología , Fosforilación , Inhibidores de Proteínas Quinasas/uso terapéutico , Ratas
14.
J Cell Biochem ; 112(4): 1103-17, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21308739

RESUMEN

In recent years many risk factors for the development of breast cancer that are linked to estrogens have been identified, and roscovitine (ROSC), a selective cyclin-dependent kinase (CDK) inhibitor, has been shown to be an efficient inhibitor of the proliferation of human breast cancer cells. Therefore, we have examined the possibility that interference with estrogen signaling pathways, using tamoxifen (TAM), a selective estrogen receptor modulator (SERM), could modulate the efficacy of treatment with ROSC. In conjunction with TAM, ROSC exhibited enhanced anti-proliferative activity and CDK inhibition, particularly in estrogen-dependent MCF-7 cells. The interaction between both drugs was synergistic. However, in ER-α-negative cells the interaction was antagonistic. Exposure of MCF-7 cells to ROSC abolished the activating phosphorylation of CDK2 and CDK7 at Ser(164/170). This in turn prevented the phosphorylation of the carboxyl-terminal repeat domain of RNA Polymerase II and ER-α at Ser(118), resulting in the down-regulation of the latter. Concomitantly, wt p53 was strongly activated by phosphorylation at Ser(46). Our results demonstrate that ROSC negatively affects the functional status of ER-α, making it potentially useful in the treatment of estrogen-dependent breast cancer cells.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Receptor alfa de Estrógeno/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Purinas/farmacología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/metabolismo , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Citometría de Flujo , Fase G2/efectos de los fármacos , Células HeLa , Humanos , Immunoblotting , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Fosforilación/efectos de los fármacos , Roscovitina , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Serina/metabolismo , Tamoxifeno/farmacología , Factores de Tiempo , Quinasa Activadora de Quinasas Ciclina-Dependientes
15.
J Cell Physiol ; 226(2): 341-9, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20836132

RESUMEN

Complexes consisting of cyclin-dependent kinases (CDKs) and their regulatory subunits (the cyclins) control the progression of normal mammalian cells through the cell cycle. However, during malignant transformation this regulatory apparatus malfunctions, allowing cells to undergo unchecked proliferation. In many cases, the high mitotic potential of malignant cells is due to the constitutive activation of CDK-cyclin complexes, facilitated by the inactivation of cellular CDK inhibitors, such as p16(INK4A) or p27(Kip1), and the loss of functional tumor suppressors, such as the p53 and pRb proteins. It has recently been suggested that pharmacological intervention based on remedying the deficiency or loss of activity of these negative regulators of the cell cycle could be a very effective therapeutic option in the treatment of cancer. Multiple CDK inhibitors have been synthesized over the last two decades, spanning at least five classes of compounds. While these inhibitors can be classified on the basis of their chemical structure, it may be more interesting to categorize them according to their pharmacological nature, as broad-spectrum unspecific, pan-specific, or very selective antagonists. This review offers a critical assessment of the advantages and disadvantages of both pan-specific and highly selective CDK inhibitors in therapy.


Asunto(s)
Quinasas Ciclina-Dependientes , Ciclinas/metabolismo , Neoplasias , Inhibidores de Proteínas Quinasas/uso terapéutico , Ciclo Celular/fisiología , Ensayos Clínicos como Asunto , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina/uso terapéutico , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/metabolismo , Activación Enzimática , Humanos , Estructura Molecular , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Inhibidores de Proteínas Quinasas/química , Subunidades de Proteína/metabolismo , Proteínas Supresoras de Tumor/metabolismo
16.
J Cell Biochem ; 112(1): 273-88, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21080333

RESUMEN

Human MCF-7 breast cancer cells are resistant to pro-apoptotic stimuli due to caspase-3 inactivation. On the other hand, they should be sensitive to agents like selective pharmacological inhibitors of cyclin-dependent kinases (CDKs) that (re)activate p53 tumor suppressor protein because they harbor intact p53 pathways. In this study we examined whether reconstitution of caspase-3 in MCF-7 cells sensitizes them to inhibitors of CDKs, by analyzing the effects of roscovitine (ROSC) and olomoucine (OLO), two closely related selective pharmacological CDK inhibitors, on both mother MCF-7 cells and a secondary mutant line, MCF-7.3.28 that stably expresses human caspase-3. The results show that ROSC is, as expected, much more potent than OLO. Surprisingly; however, ROSC and OLO reduced proliferation of parental MCF-7 cells more strongly than caspase-3-proficient counterparts. Both inhibitors arrest human breast cancer cells at the G(2)-phase of the cell cycle. Analysis of cell-cycle regulators by immunoblotting revealed that ROSC strongly induces p53 protein activity by inducing its phosphorylation at Ser46 in the MCF-7 cells lacking caspase-3, but not in caspase-3-proficient cells. Furthermore, reconstitution of caspase-3 in MCF-7 cells neither elevates the mitochondrial apoptosis rate nor significantly increases caspases activity upon ROSC treatment. However, the stabilization of p53 in response to DNA damaging agents is the same in both caspase negative and positive MCF-7 cells. Cytotoxic agents induce caspase-3-dependent apoptosis in caspase-3-proficient cells. These results indicate that reconstitution of MCF-7 cancer cells with caspase-3 sensitize them to the action of DNA damaging agents but not to ATP-like pharmacological inhibitors of CDKs.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Caspasa 3/metabolismo , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular , Quinasas Ciclina-Dependientes/metabolismo , Femenino , Humanos , Fosforilación , Inhibidores de Proteínas Quinasas/uso terapéutico
17.
Acta Biochim Pol ; 56(3): 495-501, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19724778

RESUMEN

Increased expression and activity of proteins driving cell cycle progression as well as inactivation of endogenous inhibitors of cyclin-dependent kinases (CDKs) enhance the proliferative potential of cells. Escape of cells during malignant transformation from the proper cell cycle control rendering them independent from growth factors provides rationale for therapeutic targeting of CDKs. Exposure of rapidly growing human MCF-7 breast cancer and HeLa cervix cancer cells to roscovitine (ROSC), a selective inhibitor of CDKs, inhibits their proliferation by induction of cell cycle arrest and/or apoptosis. The outcome strongly depends on the intrinsic traits of the tumor cells, on their cell cycle status prior to the onset of treatment and also on ROSC concentration. At lower dose ROSC primarily inhibits the cell cycle-related CDKs resulting in a strong cell cycle arrest. Interestingly, ROSC arrests asynchronously growing cells at the G(2)/M transition irrespective of the status of their restriction checkpoint. However, the exposure of cancer cells synchronized after serum starvation in the late G(1) phase results in a transient G(1) arrest only in cells displaying the intact G(1)/S checkpoint. At higher dosage ROSC triggers apoptosis. In HeLa cells inhibition of the activity of CDK7 and, in consequence, that of RNA polymerase II is a major event that facilitates the initiation of caspase-dependent apoptosis. In contrast, in the caspase-3-deficient MCF-7 breast cancer cells ROSC induces apoptosis by a p53-dependent pathway. HIPK2-mediated activation of the p53 transcription factor by phosphorylation at Ser46 results in upregulation of p53AIP1 protein. This protein after de novo synthesis and translocation into the mitochondria promotes depolarization of the mitochondrial membrane.


Asunto(s)
Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Purinas/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Citometría de Flujo , Fase G2/efectos de los fármacos , Células HeLa , Humanos , Immunoblotting , Fosforilación/efectos de los fármacos , Roscovitina , Proteína p53 Supresora de Tumor/metabolismo , Neoplasias del Cuello Uterino/tratamiento farmacológico
18.
Ann N Y Acad Sci ; 1171: 250-6, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19723062

RESUMEN

Roscovitine (ROSC), a selective blocker of cyclin-dependent kinases (CDKs) efficiently inhibits proliferation of exponentially growing human MCF-7 breast cancer cells by induction of cell cycle arrest and p53-mediated apoptosis. ROSC blocks MCF-7 cells in G(2) phase in a time- and concentration-dependent manner. However, ROSC exerts a much weaker antiproliferative effect on human diploid fibroblasts. Therefore, in this study we questioned whether and to what extent the antiproliferative effect of ROSC depends on the cell cycle status of cancer cells exposed to the drug. We investigated the action of ROSC on asynchronous, exponentially growing, and on synchronized human MCF-7 breast cancer cells. MCF-7 cells were arrested in G(1) phase after serum withdrawal and in S phase by hydroxyurea. After serum refeeding, synchronized cells started to reenter the active cell cycle after 12 h. Exposure of G(1)-synchronized cells to ROSC prolonged the cell cycle arrest and was accompanied by a decrease in S-phase cells after 24 h. A similar but weaker trend occurred after ROSC administration, to cells released from G(1) arrest for 4 h. ROSC diminished the frequency of S-phase cells. Exposure of MCF-7 cells released from G(1) arrest to ROSC for 24 h resulted in an increase of the G(1)-cell population by 20%. Exposure to ROSC of MCF-7 cells released from S-phase block increased the ratio of S-phase cells. These results indicate that the cell cycle status of cancer cells prior to the onset of therapy determines the outcome of treatment.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Purinas/farmacología , Antineoplásicos/farmacología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/fisiopatología , División Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Medio de Cultivo Libre de Suero/farmacología , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Citometría de Flujo , Fase G2/efectos de los fármacos , Humanos , Hidroxiurea/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Roscovitina , Fase S/efectos de los fármacos
19.
J Cell Biochem ; 106(5): 937-55, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19180585

RESUMEN

Exposure of asynchronously growing human HeLa cervical carcinoma cells to roscovitine (ROSC), a selective cyclin-dependent kinases (CDKs) inhibitor, arrests their progression at the transition between G(2)/M and/or induces apoptosis. The outcome depends on the ROSC concentration. At higher dose ROSC represses HPV-encoded E7 oncoprotein and initiates caspase-dependent apoptosis. Inhibition of the site-specific phosphorylation of survivin and Bad, occurring at high-dose ROSC treatment, precedes the onset of apoptosis and seems to be a prerequisite for cell death. Considering the fact that in HeLa cells the G(1)/S restriction checkpoint is abolished by E7, we addressed the question whether the inhibition of CDKs by pharmacological inhibitors in synchronized cells would be able to block the cell-cycle in G(1) phase. For this purpose, we attempted to synchronize cells by serum withdrawal or by blocking of the mitotic apparatus using nocodazole. Unlike human MCF-7 cells, HeLa cells do not undergo G(1) block after serum starvation, but respond with a slight increase of the ratio of G(1) population. Exposure of G(1)-enriched HeLa cells to ROSC after re-feeding does not block their cell-cycle progression at G(1)-phase, but increases the ratio of S- and G(2)-phase, thereby mimicking the effect on asynchronously growing cells. A quite different impact is observed after treatment of HeLa cells released from mitotic block. ROSC prevents their cell cycle progression and cells transiently accumulate in G(1)-phase. These results show that inhibition of CDKs by ROSC in cells lacking the G(1)/S restriction checkpoint has different outcomes depending on the cell-cycle status prior to the onset of treatment.


Asunto(s)
Ciclo Celular , Purinas/farmacología , Neoplasias del Cuello Uterino/tratamiento farmacológico , Medio de Cultivo Libre de Suero/farmacología , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Femenino , Fase G1 , Células HeLa , Humanos , Roscovitina , Neoplasias del Cuello Uterino/patología
20.
Food Chem Toxicol ; 46(4): 1327-33, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17933449

RESUMEN

Human MCF-7 breast cancer cells are relatively resistant to anti-cancer drugs. Recently, we reported that roscovitine (ROSC), a selective cyclin-dependent kinase (CDK) inhibitor, arrested human MCF-7 breast cancer cells in G2 phase of the cell cycle and concomitantly induced apoptosis. Moreover, we observed that the effect of the CDK inhibitor was dependent on the content of the culture medium. The cell cycle inhibiting action of ROSC was markedly diminished in human MCF-7 cells cultivated in medium supplemented with phenol red. These observations indicated that the therapeutic effects of ROSC can be affected by the components of the tissue medium. Recently, a number of epidemiological and experimental studies indicated that polyphenols (e.g. resveratrol, epicatechins etc.), abundant micronutrients in food, are anti-oxidant agents and could have strong anti-mitotic as well as pro-apoptotic activities. In the present contribution we raised the question whether the ROSC-mediated cell cycle arrest could be additionally modulated by compounds of natural origin, especially by polyphenols. Considering the potential benefits of the dietary components during the post-chemotherapy period, we focused our attention on the effects of resveratrol administration after treatment with ROSC. We analyzed whether the combined treatment with resveratrol would exert any additional effect on the cell cycle status of ROSC-treated human cancer cells. Resveratrol exhibited low direct cytotoxicity. The combined treatment with ROSC enhanced the ROSC-mediated inhibition of cell proliferation and cell cycle arrest. These results indicate that targeted combination of anti-cancer drugs with distinct naturally occurring compounds could increase the efficacy of the therapy and concomitantly reduce the undesired side effects exerted by cytostatic drugs.


Asunto(s)
Antineoplásicos/farmacología , Antioxidantes/farmacología , Ciclo Celular/efectos de los fármacos , Purinas/farmacología , Estilbenos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Recuento de Células , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , ADN/biosíntesis , ADN/genética , Femenino , Citometría de Flujo , Humanos , Resveratrol , Roscovitina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA