Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Br J Cancer ; 109(12): 3034-41, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24178758

RESUMEN

BACKGROUND: Persin is a plant toxin that displays synergistic cytotoxicity with tamoxifen in human breast cancer cell lines. Here, we examined the ability of persin to circumvent tamoxifen resistance and delineated the intracellular signalling pathways involved. METHODS: The induction of apoptosis in tamoxifen-resistant and -sensitive breast cancer cells was measured by flow cytometry following treatment with persin±tamoxifen. Markers of endoplasmic reticulum stress (ERS) were analysed following treatment, and their causal role in mediating persin-induced apoptosis was determined using chemical inhibitors and RNA interference. RESULTS: Cells that were resistant to an apoptotic concentration of tamoxifen maintained an apoptotic response to persin. Persin-induced apoptosis was associated with an increase in markers of ERS, that is, CHOP expression and XBP-1 splicing and was decreased by CHOP siRNA. The CASP-4 inhibitor Z-YVAD-FMK markedly inhibited persin-induced apoptosis in both tamoxifen-sensitive and -resistant cells. CONCLUSION: The cytotoxic effects of persin are CASP-4 dependent and mediated by CHOP-dependent and -independent ERS signalling cascades. Increased ERS signalling contributes to persin-induced reversal of tamoxifen resistance.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Alcoholes Grasos/farmacología , Extractos Vegetales/farmacología , Tamoxifeno/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Alcoholes Grasos/administración & dosificación , Femenino , Humanos , Células MCF-7 , Transducción de Señal , Tamoxifeno/administración & dosificación
2.
Ann Oncol ; 23(7): 1713-22, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22241899

RESUMEN

BACKGROUND: Current staging methods for pancreatic cancer (PC) are inadequate, and biomarkers to aid clinical decision making are lacking. Despite the availability of the serum marker carbohydrate antigen 19.9 (CA19.9) for over two decades, its precise role in the management of PC is yet to be defined, and as a consequence, it is not widely used. METHODS: We assessed the relationship between perioperative serum CA19.9 levels, survival and adjuvant chemotherapeutic responsiveness in a cohort of 260 patients who underwent operative resection for PC. RESULTS: By specifically assessing the subgroup of patients with detectable CA19.9, we identified potential utility at key clinical decision points. Low postoperative CA19.9 at 3 months (median survival 25.6 vs 14.8 months, P=0.0052) and before adjuvant chemotherapy were independent prognostic factors. Patients with postoperative CA 19.9 levels>90 U/ml did not benefit from adjuvant chemotherapy (P=0.7194) compared with those with a CA19.9 of ≤90 U/ml (median 26.0 vs 16.7 months, P=0.0108). Normalization of CA19.9 within 6 months of resection was also an independent favorable prognostic factor (median 29.9 vs 14.8 months, P=0.0004) and normal perioperative CA19.9 levels identified a good prognostic group, which was associated with a 5-year survival of 42%. CONCLUSIONS: Perioperative serum CA19.9 measurements are informative in patients with detectable CA19.9 (defined by serum levels of >5 U/ml) and have potential clinical utility in predicting outcome and response to adjuvant chemotherapy. Future clinical trials should prioritize incorporation of CA19.9 measurement at key decision points to prospectively validate these findings and facilitate implementation.


Asunto(s)
Biomarcadores de Tumor/sangre , Antígeno CA-19-9/sangre , Carcinoma Ductal Pancreático/sangre , Recurrencia Local de Neoplasia , Neoplasias Pancreáticas/sangre , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Ductal Pancreático/diagnóstico , Carcinoma Ductal Pancreático/mortalidad , Carcinoma Ductal Pancreático/terapia , Quimioterapia Adyuvante , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Pancreatectomía , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/terapia , Periodo Perioperatorio , Pronóstico , Modelos de Riesgos Proporcionales , Estudios Retrospectivos
3.
Br J Cancer ; 100(1): 123-33, 2009 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-19066611

RESUMEN

BAG-1 (bcl-2-associated athanogene) enhances oestrogen receptor (ER) function and may influence outcome and response to endocrine therapy in breast cancer. We determined relationships between BAG-1 expression, molecular phenotype, response to tamoxifen therapy and outcome in a cohort of breast cancer patients and its influence on tamoxifen sensitivity in MCF-7 breast cancer cells in vitro. Publically available gene expression data sets were analysed to identify relationships between BAG-1 mRNA expression and patient outcome. BAG-1 protein expression was assessed using immunohistochemistry in 292 patients with invasive ductal carcinoma and correlated with clinicopathological variables, therapeutic response and disease outcome. BAG-1-overexpressing MCF-7 cells were treated with antioestrogens to assess its effects on cell proliferation. Gene expression data demonstrated a consistent association between high BAG-1 mRNA and improved survival. In ER+ cancer (n=189), a high nuclear BAG-1 expression independently predicted improved outcome for local recurrence (P=0.0464), distant metastases (P=0.0435), death from breast cancer (P=0.009, hazards ratio 0.29, 95% CI: 0.114-0.735) and improved outcome in tamoxifen-treated patients (n=107; P=0.0191). BAG-1 overexpression in MCF-7 cells augmented antioestrogen-induced growth arrest. A high BAG-1 expression predicts improved patient outcome in ER+ breast carcinoma. This may reflect both a better definition of the hormone-responsive phenotype and a concurrent increased sensitivity to tamoxifen.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Carcinoma Ductal de Mama/tratamiento farmacológico , Proteínas de Unión al ADN/fisiología , Antagonistas de Estrógenos/uso terapéutico , Receptores de Estrógenos/análisis , Tamoxifeno/uso terapéutico , Factores de Transcripción/fisiología , Neoplasias de la Mama/química , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/química , Carcinoma Ductal de Mama/patología , Línea Celular Tumoral , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/genética , Femenino , Humanos , Inmunohistoquímica , Invasividad Neoplásica , ARN Mensajero/análisis , Factores de Transcripción/análisis , Factores de Transcripción/genética
4.
Mol Cell Biol ; 11(10): 5032-43, 1991 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-1922031

RESUMEN

This study documents a biphasic change in the rate of cell cycle progression and proliferation of T-47D human breast cancer cells treated with synthetic progestins, consisting of an initial transient acceleration in transit through G1, followed by cell cycle arrest and growth inhibition. Both components of the response were mediated via the progesterone receptor. The data are consistent with a model in which the action of progestins is to accelerate cells already progressing through G1, which are then arrested early in G1 after completing a round of replication, as are cells initially in other phases of the cell cycle. Such acceleration implies that progestins act on genes or gene products which are rate limiting for cell cycle progression. Increased production of epidermal growth factor and transforming growth factor alpha, putative autocrine growth factors in breast cancer cells, does not appear to account for the initial response to progestins, since although the mRNA abundance for these growth factors is rapidly induced by progestins, cells treated with epidermal growth factor or transforming growth factor alpha did not enter S phase until 5 to 6 h later than those stimulated by progestin. The proto-oncogenes c-fos and c-myc were rapidly but transiently induced by progestin treatment, paralleling the well-known response of these genes to mitogenic signals in other cell types. The progestin antagonist RU 486 inhibited progestin regulation of both cell cycle progression and c-myc expression, suggesting that this proto-oncogene may participate in growth modulation by progestins.


Asunto(s)
Neoplasias de la Mama/patología , Ciclo Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Pregnenodionas/farmacología , Congéneres de la Progesterona/farmacología , Northern Blotting , Factor de Crecimiento Epidérmico/biosíntesis , Factor de Crecimiento Epidérmico/genética , Receptores ErbB/biosíntesis , Receptores ErbB/genética , Genes myc , Humanos , Cinética , Mifepristona/farmacología , Pregnenodionas/antagonistas & inhibidores , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-fos/biosíntesis , Proteínas Proto-Oncogénicas c-fos/genética , Factor de Crecimiento Transformador alfa/biosíntesis , Factor de Crecimiento Transformador alfa/genética , Células Tumorales Cultivadas
5.
Mol Cell Biol ; 20(7): 2581-91, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10713180

RESUMEN

The steroid hormone progesterone regulates proliferation and differentiation in the mammary gland and uterus by cell cycle phase-specific actions. The long-term effect of progestins on T-47D breast cancer cells is inhibition of cellular proliferation. This is accompanied by decreased G(1) cyclin-dependent kinase (CDK) activities, redistribution of the CDK inhibitor p27(Kip1) among these CDK complexes, and alterations in the elution profile of cyclin E-Cdk2 upon gel filtration chromatography, such that high-molecular-weight complexes predominate. This study aimed to determine the relative contribution of CDK inhibitors to these events. Following progestin treatment, the majority of cyclin E- and D-CDK complexes were bound to p27(Kip1) and few were bound to p21(Cip1). In vitro, recombinant His(6)-p27 could quantitatively reproduce the effects on cyclin E-Cdk2 kinase activity and the shift in molecular weight observed following progestin treatment. In contrast, cyclin D-Cdk4 was not inhibited by His(6)-p27 in vitro or p27(Kip1) in vivo. However, an increase in the expression of the Cdk4/6 inhibitor p18(INK4c) and its extensive association with Cdk4 and Cdk6 were apparent following progestin treatment. Recombinant p18(INK4c) led to the reassortment of cyclin-CDK-CDK inhibitor complexes in vitro, with consequent decrease in cyclin E-Cdk2 activity. These results suggest a concerted model of progestin action whereby p27(Kip1) and p18(INK4c) cooperate to inhibit cyclin E-Cdk2 and Cdk4. Since similar models have been developed for growth inhibition by transforming growth factor beta and during adipogenesis, interaction between the Cip/Kip and INK4 families of inhibitors may be a common theme in physiological growth arrest and differentiation.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Ciclo Celular/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/metabolismo , Progestinas/farmacología , Proteínas Supresoras de Tumor , Inhibidor p18 de las Quinasas Dependientes de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Inhibidores Enzimáticos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Pregnenodionas/farmacología , ARN Mensajero/metabolismo , Proteínas Recombinantes/metabolismo , Células Tumorales Cultivadas
6.
Mol Cell Biol ; 18(4): 1812-25, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9528753

RESUMEN

The steroid hormone progesterone regulates proliferation and differentiation in the mammary gland and uterus by cell cycle phase-specific actions. In breast cancer cells the predominant effect of synthetic progestins is long-term growth inhibition and arrest in G1 phase. Progestin-mediated growth arrest of T-47D breast cancer cells was preceded by inhibition of cyclin D1-Cdk4, cyclin D3-Cdk4, and cyclin E-Cdk2 kinase activities in vitro and reduced phosphorylation of pRB and p107. This was accompanied by decreases in the expression of cyclins D1, D3, and E, decreased abundance of cyclin D1- and cyclin D3-Cdk4 complexes, increased association of the cyclin-dependent kinase (CDK) inhibitor p27 with the remaining Cdk4 complexes, and changes in the molecular masses and compositions of cyclin E complexes. In control cells cyclin E eluted from Superdex 200 as two peaks of approximately 120 and approximately 200 kDa, with the 120-kDa peak displaying greater cyclin E-associated kinase activity. Following progestin treatment, almost all of the cyclin E was in the 200-kDa, low-activity form, which was associated with the CDK inhibitors p21 and p27; this change preceded the inhibition of cell cycle progression. These data suggest preferential formation of this higher-molecular-weight, CDK inhibitor-bound form and a reduced number of cyclin E-Cdk2 complexes as mechanisms for the decreased cyclin E-associated kinase activity following progestin treatment. Ectopic expression of cyclin D1 in progestin-inhibited cells led to the reappearance of the 120-kDa active form of cyclin E-Cdk2 preceding the resumption of cell cycle progression. Thus, decreased cyclin expression and consequent increased CDK inhibitor association are likely to mediate the decreases in CDK activity accompanying progestin-mediated growth inhibition.


Asunto(s)
Neoplasias de la Mama/enzimología , Quinasas CDC2-CDC28 , Proteínas de Ciclo Celular , Ciclina D1/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Progestinas/farmacología , Proteínas Proto-Oncogénicas , Proteínas Supresoras de Tumor , Neoplasias de la Mama/metabolismo , Ciclina D3 , Ciclina E/metabolismo , Ciclina G , Ciclina G1 , Quinasa 2 Dependiente de la Ciclina , Quinasa 4 Dependiente de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Inhibidores Enzimáticos/metabolismo , Femenino , Humanos , Proteínas Asociadas a Microtúbulos/metabolismo , Peso Molecular , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína de Retinoblastoma/metabolismo , Proteína p107 Similar a la del Retinoblastoma , Células Tumorales Cultivadas
7.
Mol Cell Biol ; 18(8): 4499-508, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9671459

RESUMEN

Estrogen-induced progression through G1 phase of the cell cycle is preceded by increased expression of the G1-phase regulatory proteins c-Myc and cyclin D1. To investigate the potential contribution of these proteins to estrogen action, we derived clonal MCF-7 breast cancer cell lines in which c-Myc or cyclin D1 was expressed under the control of the metal-inducible metallothionein promoter. Inducible expression of either c-Myc or cyclin D1 was sufficient for S-phase entry in cells previously arrested in G1 phase by pretreatment with ICI 182780, a potent estrogen antagonist. c-Myc expression was not accompanied by increased cyclin D1 expression or Cdk4 activation, nor was cyclin D1 induction accompanied by increases in c-Myc. Expression of c-Myc or cyclin D1 was sufficient to activate cyclin E-Cdk2 by promoting the formation of high-molecular-weight complexes lacking the cyclin-dependent kinase inhibitor p21, as has been described, following estrogen treatment. Interestingly, this was accompanied by an association between active cyclin E-Cdk2 complexes and hyperphosphorylated p130, identifying a previously undefined role for p130 in estrogen action. These data provide evidence for distinct c-Myc and cyclin D1 pathways in estrogen-induced mitogenesis which converge on or prior to the formation of active cyclin E-Cdk2-p130 complexes and loss of inactive cyclin E-Cdk2-p21 complexes, indicating a physiologically relevant role for the cyclin E binding motifs shared by p130 and p21.


Asunto(s)
Quinasas CDC2-CDC28 , Ciclo Celular , Ciclina D1/metabolismo , Ciclina E/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Estradiol/metabolismo , Estrógenos/metabolismo , Imitación Molecular , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas , Ciclina D1/genética , Quinasa 2 Dependiente de la Ciclina , Quinasa 4 Dependiente de la Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Estradiol/análogos & derivados , Estradiol/farmacología , Antagonistas de Estrógenos/farmacología , Fulvestrant , Fase G1 , Humanos , Fosfoproteínas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-myc/genética , Purinas/farmacología , Proteínas Recombinantes de Fusión/metabolismo , Proteína p130 Similar a la del Retinoblastoma , Roscovitina , Fase S , Células Tumorales Cultivadas
8.
Mol Cell Biol ; 13(6): 3577-87, 1993 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-8497271

RESUMEN

Cyclins and proto-oncogenes including c-myc have been implicated in eukaryotic cell cycle control. The role of cyclins in steroidal regulation of cell proliferation is unknown, but a role for c-myc has been suggested. This study investigated the relationship between regulation of T-47D breast cancer cell cycle progression, particularly by steroids and their antagonists, and changes in the levels of expression of these genes. Sequential induction of cyclins D1 (early G1 phase), D3, E, A (late G1-early S phase), and B1 (G2 phase) was observed following insulin stimulation of cell cycle progression in serum-free medium. Transient acceleration of G1-phase cells by progestin was also accompanied by rapid induction of cyclin D1, apparent within 2 h. This early induction of cyclin D1 and the ability of delayed administration of antiprogestin to antagonize progestin-induced increases in both cyclin D1 mRNA and the proportion of cells in S phase support a central role for cyclin D1 in mediating the mitogenic response in T-47D cells. Compatible with this hypothesis, antiestrogen treatment reduced the expression of cyclin D1 approximately 8 h before changes in cell cycle phase distribution accompanying growth inhibition. In the absence of progestin, antiprogestin treatment inhibited T-47D cell cycle progression but in contrast did not decrease cyclin D1 expression. Thus, changes in cyclin D1 gene expression are often, but not invariably, associated with changes in the rate of T-47D breast cancer cell cycle progression. However, both antiestrogen and antiprogestin depleted c-myc mRNA by > 80% within 2 h. These data suggest the involvement of both cyclin D1 and c-myc in the steroidal control of breast cancer cell cycle progression.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Quinasas CDC2-CDC28 , Ciclo Celular/fisiología , Quinasas Ciclina-Dependientes , Ciclinas/genética , Estradiol/análogos & derivados , Regulación Neoplásica de la Expresión Génica/fisiología , Sustancias de Crecimiento/farmacología , Mifepristona/farmacología , Pregnenodionas/farmacología , Proteínas Serina-Treonina Quinasas , Proteína Quinasa CDC2/biosíntesis , Proteína Quinasa CDC2/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Quinasa 2 Dependiente de la Ciclina , Ciclinas/biosíntesis , ADN de Neoplasias/análisis , ADN de Neoplasias/genética , Estradiol/farmacología , Antagonistas de Estrógenos/farmacología , Femenino , Fase G1 , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes myc , Histonas/genética , Humanos , Insulina/farmacología , Cinética , Alcamidas Poliinsaturadas , Congéneres de la Progesterona/farmacología , Proteínas Quinasas/biosíntesis , Proteínas Quinasas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/farmacología , Factores de Tiempo , Transcripción Genética/efectos de los fármacos , Células Tumorales Cultivadas
9.
Cancer Res ; 48(21): 6037-43, 1988 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-3167854

RESUMEN

In an attempt to elucidate the role of somatic mutation in the development of resistance to cancer chemotherapy, an assay was sought to measure the frequency of hypoxanthine-guanine phosphoribosyltransferase (HPRT) mutants in human tumors. Based on the same principle as [3H]thymidine/autoradiography, a method was developed to identify cell proliferation using the thymidine analog 5-bromo-2'-deoxyuridine (BrdUrd). BrdUrd incorporation into DNA was measured following the immunofluorescent staining of fixed cells using a monoclonal antibody highly specific for this nucleoside analog. The human leukemic cell line, CCRF-CEM, was used to investigate the conditions necessary for the stringent selection of HPRT- mutants using 6-thioguanine (6TG). The appropriate 6TG exposure necessary to inhibit BrdUrd incorporation in wild-type cells, while allowing proliferation of spontaneous HPRT- mutants, was greater than or equal to 30 microM 6TG for 72 h (10 microM BrdUrd added 24 h prior to harvest). BrdUrd did not affect the growth of HPRT- mutants in the presence of 6TG. BrdUrd-labeled 6TG-resistant cells were enumerated flow cytometrically using fluorescent microspheres as an internal standard and the nonparametric, Kolmogorov-Smirnov test was used for independent statistical analysis of the subpopulations of fluorescent, 6TG-resistant cells. Evidence that CCRF-CEM cells which incorporated BrdUrd in the presence of 6TG were, in fact, HPRT- mutants was sought. It was demonstrated that spontaneous 6TG-resistant cells from the CCRF-CEM population were reduced by growth in medium containing aminopterin. The mutant frequency in the CCRF-CEM cell line was found to be 4.28 x 10(-5) +/- 2.04 x 10(-5) using the BrdUrd/flow cytometric technique.


Asunto(s)
Citometría de Flujo , Mutación , Tioguanina/farmacología , Bromodesoxiuridina/metabolismo , Medios de Cultivo , Resistencia a Medicamentos , Humanos , Hipoxantina Fosforribosiltransferasa/genética , Células Tumorales Cultivadas/efectos de los fármacos
10.
Cancer Res ; 50(16): 4849-55, 1990 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-2379149

RESUMEN

A previous study from this laboratory (Koga et al., Cancer Res., 48: 2734-2739, 1988) demonstrated that the growth inhibitory effect of 1,25-dihydroxyvitamin D3 in human breast cancer cells in vitro was associated with a decline in the concentration of epidermal growth factor receptor (EGF-R). In the present study experiments were undertaken with the phorbol ester, 12-O-tetradecanoyl-phorbol-13-acetate (TPA), an agent known to decrease EGF-R binding, in order to further define the relationship between changes in EGF-R binding and changes in growth rates in 10 human breast cancer cell lines. Treatment with TPA decreased the rate of cell proliferation in all cell lines except BT 474 in which a slight increase in proliferation rate was observed. Sensitivity to TPA was unrelated to estrogen receptor (ER) status and a wide spectrum of sensitivities was apparent. The concentrations of TPA that produced a 25% decrease in cell number ranged from less than 0.25 nM for MCF 7M and BT 20 cells to greater than 10 nM for the HBL 100, T 47D, and ZR 75-1 cell lines. Growth inhibition was associated with a block in cell cycle progression in the G1 and G2 + M phases of the cell cycle. In all cell lines studied, except BT 474, TPA treatment resulted in a reduction in the ability of cells to bind EGF. Saturation analysis revealed marked differences between the effects of TPA on EGF binding in ER+ and ER- cell lines. In ER+ cell lines, 2-h treatment with 10 nM TPA resulted in a marked reduction in the number of high affinity EGF-R sites and a significant decrease in binding affinity. Among this group of cell lines there was a significant positive correlation between the ability of TPA to decrease cell growth and the TPA-induced decrease in the number of EGF-R sites/cell (r = 0.82, P less than 0.03). In ER- cell lines, TPA-induced growth inhibition and the minor changes in EGF-R concentration were unrelated. However, growth inhibition was negatively correlated with TPA-induced changes in apparent affinity of the EGF-R (r = 1.00, P less than 0.003) and in the same rank order as the EGF-R concentration in control cells. These data demonstrate differential relationships between TPA-induced changes in growth and EGF-R binding in ER+ and ER- breast cancer cells, thus supporting the view that growth regulatory pathways are markedly different in these two subtypes of human breast cancer.


Asunto(s)
Proteínas de Caenorhabditis elegans , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/fisiología , Proteína Quinasa C , Receptores de Droga/fisiología , Receptores de Estrógenos/fisiología , Acetato de Tetradecanoilforbol/farmacología , Células Tumorales Cultivadas/citología , Neoplasias de la Mama , Proteínas Portadoras , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular , Femenino , Humanos , Cinética , Receptores de Estrógenos/análisis , Células Tumorales Cultivadas/efectos de los fármacos
11.
Cancer Res ; 49(1): 112-6, 1989 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-2642286

RESUMEN

The molecular basis of the growth-inhibitory effects of progestins or antiestrogens in human breast cancer has not been fully elucidated. Both direct actions and indirect actions, where the growth inhibition results from modulation of the production of, and/or the response to, growth factors, have been proposed. In this study the ability of some growth factors to modulate progestin-induced inhibition of cell proliferation was investigated in vitro, using T-47D human breast cancer cells. When T-47D cells grown in insulin-containing medium were treated for 4 to 5 days with the synthetic progestin, ORG 2058, at a concentration of 10 nM, cell numbers were reduced to 10 to 20% of control. Simultaneous treatment with epidermal growth factor (EGF) and ORG 2058 led to a partial reversal of the growth-inhibitory effect of the progestin. The magnitude of the effect of EGF was concentration dependent, being half-maximal at 0.48 ng/ml (0.08 nM) and maximal at concentrations greater than 5 ng/ml (greater than 0.8 nM), where cell numbers were increased by 50% compared to those in the presence of ORG 2058 alone. ORG 2058 was no more potent in the absence of insulin, and, after several passages in insulin-free medium, addition of insulin failed to modulate the effect of ORG 2058. However, when maximal concentrations of insulin (5 micrograms/ml) and EGF (10 ng/ml) were administered together with ORG 2058, insulin and EGF appeared to act synergistically to reduce the ORG 2058-induced inhibition of proliferation. In similar experiments in which cells were treated with hydroxyclomiphene, a potent antiestrogen, insulin was shown to partially reverse the growth-inhibitory effects of hydroxyclomiphene. Significant increases in cell number above hydroxyclomiphene-treated controls were apparent at insulin concentrations greater than 50 ng/ml, and at 5 micrograms/ml the increase was approximately 2-fold. In contrast to the situation with progestins, simultaneous treatment with EGF and insulin had only an additive effect in reversing the growth-inhibitory effect of the antiestrogen. The results are compatible with the hypothesis that part of the growth-inhibitory effects of progestin and antiestrogen on human breast cancer cell proliferation is mediated by inhibition of autocrine growth factor production. However, they do not exclude more direct mechanisms involving modulation of progesterone and/or estrogen receptors by EGF and/or insulin.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Neoplasias de la Mama/patología , Clomifeno/análogos & derivados , Factor de Crecimiento Epidérmico/farmacología , Antagonistas de Estrógenos/farmacología , Inhibidores de Crecimiento/farmacología , Insulina/farmacología , Progestinas/farmacología , División Celular/efectos de los fármacos , Clomifeno/farmacología , Femenino , Humanos , Pregnenodionas/farmacología , Células Tumorales Cultivadas
12.
Cancer Res ; 49(9): 2398-404, 1989 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-2706627

RESUMEN

Tamoxifen and other structurally related nonsteroidal antiestrogens possess properties in addition to their estrogen antagonist activity including inhibition of both calmodulin and protein kinase C. The present studies were designed to test whether the estrogen-reversible (estrogen receptor mediated) and estrogen-irreversible effects of nonsteroidal antiestrogens on cell cycle progression in vitro were mediated at the same or different points within the cell cycle and if the estrogen-irreversible effects coincided temporally with that of a calmodulin antagonist, R24571. Initial experiments investigated the effects of ICI 164384, a pure estrogen antagonist, on proliferation kinetics in asynchronous cultures of MCF-7 human breast cancer cells. At concentrations greater than 1 nM ICI 164384 significantly reduced growth rate while at greater than or equal to 50 nM, ICI 164384 completely arrested growth after the first 24 h of exposure. Concentrations up to 5 microM failed either to cause more profound effects on growth or induce cytotoxicity. Growth inhibition was associated with a decrease in the proportion of S phase cells and an accumulation of cells in G1 phase, and was completely reversed by the simultaneous addition of equimolar estradiol. In order to identify the points of action within the cell cycle of ICI 164384, and the estrogen-reversible and estrogen-irreversible components of the nonsteroidal estrogen antagonist, hydroxyclomiphene, and the calmodulin antagonist, R24571, experiments were undertaken with MCF-7 cells synchronized by mitotic selection. The mean point of action was assessed by delaying addition of the drugs for increasing time periods following mitotic selection and using DNA flow cytometry to determine the proportion of the population affected by drug administration at a specific time within G1 phase. These studies showed that sensitivity to ICI 164384 was restricted to the early part of G1 phase and that the mean time of action was 4.9 h after the beginning of G1 for this pure estrogen antagonist. The mean times of action of the estrogen-reversible (4.1 h into G1 phase) and estrogen-irreversible (4.1 h) mechanisms of action of hydroxyclomiphene, and R24571 (4.0 h), all appeared to be within a similar time frame in early to mid G1 phase. It is concluded that ICI 164384 inhibits breast cancer cell proliferation by inducing a transition delay in G1 phase and that the point of action of this pure estrogen antagonist in early G1 phase is indistinguishable temporally from that of nonsteroidal antiestrogens and calmodulin antagonists.


Asunto(s)
Neoplasias de la Mama/patología , Calmodulina/antagonistas & inhibidores , Ciclo Celular/efectos de los fármacos , Antagonistas de Estrógenos/farmacología , Clomifeno/análogos & derivados , Clomifeno/farmacología , ADN/análisis , Estradiol/análogos & derivados , Estradiol/farmacología , Femenino , Humanos , Imidazoles/farmacología , Alcamidas Poliinsaturadas , Células Tumorales Cultivadas
13.
Cancer Res ; 48(18): 5151-6, 1988 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-3044582

RESUMEN

Ribonucleotide reductase catalyzes the first unique, rate-limiting step in DNA synthesis; both its large (M1) and small (M2) subunits are necessary for activity. While direct studies of M2 expression have previously shown a tight correlation with S phase, the kinetic features of M1-expressing cells have remained ill defined. Therefore we have, using immunofluorescence flow cytometry, analyzed changes in whole cell M1 levels and DNA content during various cell cycle and differentiation events. In asynchronous cultures M1 levels are sustained throughout the cell cycle, including G1 phase when M2 levels and ribonucleotide reductase catalytic activity are known to be very low. In contrast M1 is virtually absent from quiescent lymphocytes but is expressed following mitogen stimulation, shortly before S phase cells appear. M1 declines to low levels in "plateau phase" cultures, the major reduction occurring in cells with 2n (G0/G1) DNA content. HL-60 promyelocytic leukemia cells, induced into either myeloid or monocyte-macrophage differentiation, show a similar marked decrease in M1 levels concomitant with the cessation of cell division. We conclude that the M1 subunit of ribonucleotide reductase is constitutively expressed by cycling cells. It is acquired during stimulated transition from G0 to G1 and is lost during exit to G0 or terminal differentiation. This pattern of expression suggests that determination of cellular M1 content may be useful in distinguishing proliferating (including G1) and quiescent (including G0) cells in vivo.


Asunto(s)
Leucemia Mieloide Aguda/enzimología , Ribonucleótido Reductasas/metabolismo , Diferenciación Celular , División Celular , Línea Celular , Supervivencia Celular , Humanos , Interfase , Sustancias Macromoleculares , Acetato de Tetradecanoilforbol/farmacología
14.
Cancer Res ; 48(18): 5084-91, 1988 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-2970295

RESUMEN

The effect of medroxyprogesterone acetate (MPA) on breast cancer cell proliferation kinetics was investigated in ten human breast cell lines growing as monolayer cultures. Significant inhibition of growth occurred only in the estrogen receptor-positive, progesterone receptor-positive cell lines, T-47D, MCF-7, ZR 75-1, BT 474, and MDA-MB-361. Among these cell lines sensitivity to MPA varied widely; concentrations required for 20% inhibition of growth ranged from 0.04 nM for T-47D to greater than 100 nM for ZR 75-1 cells. Furthermore, although the most sensitive line, T-47D, had the highest level of PR, sensitivity to MPA was not correlated with PR levels among the responsive cell lines. More detailed studies were undertaken with the T-47D cell line. The growth-inhibitory response was confined to the progestins: MPA, ORG 2058, R5020, and progesterone, while androgens, estrogens, and glucocorticoids were without effect over the same concentration range (0.1-100 nM). MPA-induced growth inhibition was associated with a significant decrease in the proportion of S-phase cells with an accumulation of cells in the G0-G1 phase of the cell cycle. Cells began to accumulate in G0-G1 after 12 h of drug treatment and the effect was maximal by 24 h, i.e., maximal effects were observed during the first cell cycle following drug treatment. By contrast, significant accumulation in G0-G1 required exposure of MCF-7 cells to MPA for at least two cell cycle times, i.e., 48 h and the effect was still increasing at 96 h. Stathmokinetic studies revealed that in both cell lines accumulation in the G0-G1 phase was due to an MPA-induced increase in the G1 transit time. These data indicate that MPA and other progestins have direct growth inhibitory effects on estrogen receptor-positive and progesterone receptor-positive human breast cancer cells in vitro and these effects can be accounted for by a decrease in the rate at which cells traverse the G1 phase of the cell cycle.


Asunto(s)
Neoplasias de la Mama/patología , Medroxiprogesterona/análogos & derivados , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular , Estradiol/farmacología , Femenino , Citometría de Flujo , Humanos , Medroxiprogesterona/farmacología , Acetato de Medroxiprogesterona , Receptores de Progesterona/metabolismo
15.
Oncogene ; 14(11): 1329-40, 1997 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-9178893

RESUMEN

To investigate the possibility of differing roles for cyclins D1 and D2 in breast epithelial cells, we examined the expression, cell cycle regulation and activity of these two G1 cyclins in both 184 normal breast epithelial cells and T-47D breast cancer cells. Synchronisation studies in 184 cells demonstrated that cyclin D1 and cyclin D2 were differentially regulated during G1, with cyclin D2 abundance increasing by 3.7-fold but only small changes in cyclin D1 abundance observed. The functional consequences of increased cyclin D2 expression were examined in T-47D cells, which express no detectable cyclin D2. Induced expression of cyclin D2 resulted in increases in cyclin E expression, pRB phosphorylation and the percentage of cells in S-phase, while constitutive expression resulted in a consistent trend toward reduced dependence on serum for continued proliferation. Thus, cyclin D2 is a positive regulator of G1 progression in breast cells analogous to the well-documented effects of cyclin D1. Indeed, equimolar concentrations of inducible cyclin D1 and D2 resulted in quantitatively similar cell cycle effects. Marked divergence was found, however, in the CDKs activated by the two cyclins in breast epithelial cells. Cyclin D2 complexes contained a higher Cdk2/Cdk4 ratio than cyclin D1 complexes. The cyclin D2-associated kinase activity was largely inhibited by Cdk2-specific inhibitors and could phosphorylate histone H1, a substrate for Cdk2 but not for Cdk4 and Cdk6. Therefore, cyclin D2 preferentially activated Cdk2 in breast epithelial cells. In contrast, Cdk4 and Cdk6 were predominantly responsible for cyclin D1-associated kinase activity as previously reported. Thus, although cyclins D1 and D2 elicited similar effects on breast epithelial cell cycle progression they appeared to achieve this end via activation of different CDKs. This is the first evidence of cyclin D2 activating Cdk2 in mammalian cells thus providing further evidence that D-type cyclins are not necessarily redundant.


Asunto(s)
Mama/enzimología , Quinasas CDC2-CDC28 , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas , Mama/citología , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Ciclina D2 , Quinasa 2 Dependiente de la Ciclina , Quinasa 4 Dependiente de la Ciclina , Ciclinas/biosíntesis , Activación Enzimática , Células Epiteliales , Epitelio/enzimología , Humanos , Fosforilación , Unión Proteica , Proteína de Retinoblastoma/metabolismo , Fase S , Células Tumorales Cultivadas
16.
Oncogene ; 16(22): 2865-78, 1998 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-9671407

RESUMEN

The G1 cyclins, cyclin D1 and E, are rate limiting for progression through G1 phase of the cell cycle in breast epithelial cells and are oncogenic when expressed in the mammary epithelium of transgenic mice. These genes are frequently overexpressed in clinical breast cancer where overexpression appears to be associated with specific disease phenotypes, altered responsiveness to therapeutic intervention and patient survival. In order to investigate the functional correlates of cyclin D1 and cyclin E overexpression we employed a panel of normal, immortalized and neoplastic breast epithelial cell lines to examine the relationships between cyclin gene expression, cyclin-CDK complex formation and CDK activity. In agreement with earlier studies cyclin D1 and E expression varied over an approximately tenfold range among the 18 cell lines studied. There was no apparent relationship, however, between cyclin D1 expression and the in vitro activity of its major kinase partner, Cdk4, although MDA-MB-134 cells displayed the highest level of both cyclin D1 expression and Cdk4 activity. Similarly, there was no significant relationship between cyclin E expression and cyclin E-Cdk2 activity. Fractionation of whole cell lysates by gel filtration chromatography revealed that approximately 90% of the cyclin E protein was present in inactive complexes containing the CDK inhibitors p21 and p27. Much of the small fraction of active cyclin E protein was of very high apparent molecular mass, >400 kDa, suggesting that formation of these complexes is a more important determinant of cyclin E-Cdk2 activity than cyclin E abundance. These data suggest that properties of cyclins D1 and E in addition to their ability to activate Cdk4 and Cdk2 may contribute to the effects of overexpression on the breast cancer phenotype.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas de Ciclo Celular , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/biosíntesis , Proteínas Proto-Oncogénicas , Proteínas Supresoras de Tumor , Neoplasias de la Mama/enzimología , División Celular , Cromatografía en Gel , Ciclina D1/biosíntesis , Ciclina E/biosíntesis , Ciclina G , Ciclina G1 , Quinasa 4 Dependiente de la Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/biosíntesis , Inhibidores Enzimáticos/metabolismo , Células Epiteliales , Humanos , Proteínas Asociadas a Microtúbulos/biosíntesis , Células Tumorales Cultivadas
17.
Oncogene ; 15(13): 1617-23, 1997 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-9380415

RESUMEN

Chromosome 11q13 is amplified in about 13% of primary breast cancers. CCND1, encoding the cell cycle regulatory gene cyclin D1, and EMS1, encoding a filamentous actin binding protein, are favoured candidate onocogenes, whereas INT-2 is an unexpressed gene at this locus. In this study we tested the possibility that different regions of this large amplicon could be independently amplified and subsequently defined the phenotype of EMS1 amplified tumours in a series of 961 primary breast carcinomas. Using DNA slot blots, EMS1 was amplified in 15.2% of samples: 5.4% were coamplified for CCND1; 7.9% coamplified for INT-2 and 6.7% showed EMS1 amplification alone. The degree of amplification of CCND1 and INT-2 was highly correlated (P =0.0001). In contrast, no such relationship existed between EMS1 and CCND1 or INT-2 amplification, demonstrating independent amplification of EMS1 in 44% of amplified tumours. EMS1 amplification (> or = twofold increase in copy number) was positively correlated with patient age > or = 50 years (P = 0.025), ER positivity (P = 0.022), PgR positivity (P = 0.018), and was negatively correlated with HER-2/neu (c-erbB2) amplification (P = 0.01). In common with CCND1/INT-2, EMS1 amplification was associated with increased risk of relapse in patients with lymph node-negative disease (P = 0.028). In contrast, EMS1 and CCND1/INT-2 amplification appeared to confer different phenotypes in ER positive and negative tumours. A > or = threefold increase in EMS1 copy number was associated with an apparent increased risk of relapse and death in patients with ER negative tumours, but was without effect in ER positive tumours. In contrast, CCND1/INT-2 amplification had no effect in the patients with ER negative tumours but was associated with early relapse in ER positive patients. Thus EMS1 amplification may identify subgroups of breast cancer patients with increased probability of relapse and death distinct from those identified by CCND1/INT-2 amplification. Further studies are required to more clearly determine the functional consequences of EMS1 overexpression and a biological basis for the relationship between EMS1 amplification and phenotype in breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Cromosomas Humanos Par 11 , Amplificación de Genes , Proteínas de Microfilamentos , Proteínas de Neoplasias/genética , Cortactina , Ciclina D1/metabolismo , Factor 3 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/metabolismo , Marcadores Genéticos , Humanos , Fenotipo , Proteínas Proto-Oncogénicas/metabolismo
18.
Oncogene ; 11(5): 971-6, 1995 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-7675456

RESUMEN

Involvement of the retinoblastoma susceptibility (RB-1), p16INK4, p53 and telomerase genes in immortalisation was examined by determining their status in 15 human cell lines representing four immortalisation complementation groups. No abnormalities of RB-1, p53 and p16INK4 were detected in cell lines containing DNA tumour virus proteins known to bind to the protein products of the RB-1 and p53 genes. In contrast, in all other cell lines from each of the four groups either RB-1 was mutant or p16INK4 protein was undetectable and there were cell lines containing p53 mutations in three of the groups. Telomerase activity was detected in 12/15 lines, including some of the virally immortalised lines and in some lines from each group. Since none of these changes correlated with complementation group, other genetic changes must be required for immortalisation.


Asunto(s)
Proteínas Portadoras/genética , Transformación Celular Neoplásica , ADN Nucleotidilexotransferasa/genética , Genes de Retinoblastoma , Genes Supresores de Tumor , Genes p53 , Inhibidores de Proteínas Quinasas , Secuencia de Bases , Línea Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Humanos , Datos de Secuencia Molecular
19.
Oncogene ; 16(21): 2803-13, 1998 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-9652748

RESUMEN

Members of the erbB family of receptor tyrosine kinases are commonly overexpressed in human breast cancer. However, the relative contribution of particular signalling pathways activated downstream of these receptors to the mitogenic response of transformed breast epithelial cells remains poorly characterized. Administration of heregulin-beta2 (HRG), a ligand for erbB3 and erbB4, to growth arrested T-47D human breast cancer cells leads to activation of both the PI3-kinase and MAP kinase signalling pathways and potent stimulation of cell cycle progression. Specific inhibitors were used to assess the role of these pathways in HRG-induced mitogenesis and to identify underlying mechanisms in terms of regulation of gene expression. Treatment with the MEK inhibitor PD98059 led to a complete block of HRG-induced entry into S-phase, whilst administration of the PI3-kinase inhibitor wortmannin resulted in only modest inhibition. In addition, administration of PD98059 8 h after HRG was equipotent with simultaneous administration in inhibiting entry into S-phase. However, delaying addition for 14-16 h after HRG, when the cells were entering S-phase, was without effect. HRG stimulation led to sequential induction of c-myc, cyclin D1, cyclin E and cyclin A gene expression and hyperphosphorylation of the retinoblastoma protein pRB. p21 (WAF1/CIP1/SDI1) gene expression was rapidly induced by HRG, but significant changes in p27 (KIP1) protein levels were not detected. Preincubation with PD98059 blocked the HRG-dependent induction of cyclin D1 mRNA, p21 and c-Myc protein and pRB phosphorylation. These findings demonstrate that MEK activation is critical to HRG-induced S-phase entry in these cells whilst PI3-kinase plays a minor role. Moreover, these data are compatible with HRG-induced activation of MEK being critical for a mid-G1 transition point and implicate c-myc and cyclin D1 as key targets of the MAP kinase pathway involved in this response.


Asunto(s)
Neoplasias de la Mama/patología , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Proteínas Portadoras/farmacología , Glicoproteínas/farmacología , Neurregulina-1 , Androstadienos/farmacología , Neoplasias de la Mama/enzimología , Ciclo Celular/efectos de los fármacos , Ciclina D1/genética , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Ciclinas/genética , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Femenino , Flavonoides/farmacología , Expresión Génica , Humanos , Quinasas de Proteína Quinasa Activadas por Mitógenos , Mitógenos/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Inhibidores de Proteínas Quinasas , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Mensajero , Proteína de Retinoblastoma/metabolismo , Células Tumorales Cultivadas , Regulación hacia Arriba , Wortmanina
20.
Oncogene ; 8(8): 2127-33, 1993 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-8336939

RESUMEN

Cyclins, the regulatory subunits of cyclin-dependent kinases, play an important role in the control of cellular proliferation. Since dysregulated expression of these genes may contribute to the malignant phenotype the expression and amplification of cyclin A, B1, C, D1, D2, D3 and E genes were studied in 20 breast cancer cell lines. Increased expression of one or more of the cyclin A, B1, D1 or E genes was found in seven cell lines (35%); of these five (25%) showed increased expression of cyclin D1. Overexpression occurred in both the presence and absence of gene amplification. Conversely, amplification did not invariably lead to overexpression. Cyclin D2 expression was lower in breast cancer cell lines than in cultured normal breast epithelial cells. Cyclin D1 expression was further investigated in breast tumour biopsies: 56 of 124 specimens (45%) expressed higher levels of cyclin D1 mRNA than normal breast tissue. These data implicate dysregulated expression of several cyclin genes, particularly cyclin D1, as a potential factor in the pathogenesis of breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Ciclinas/genética , Amplificación de Genes , Regulación Neoplásica de la Expresión Génica , Secuencia de Bases , Femenino , Humanos , Datos de Secuencia Molecular , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA