Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell Biol ; 18(8): 4499-508, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9671459

RESUMO

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.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Ciclo Celular , Ciclina D1/metabolismo , Ciclina E/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Estradiol/metabolismo , Estrogênios/metabolismo , Mimetismo Molecular , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Proto-Oncogênicas , Ciclina D1/genética , Quinase 2 Dependente de Ciclina , Quinase 4 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Estradiol/análogos & derivados , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Fulvestranto , Fase G1 , Humanos , Fosfoproteínas/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-myc/genética , Purinas/farmacologia , Proteínas Recombinantes de Fusão/metabolismo , Proteína p130 Retinoblastoma-Like , Roscovitina , Fase S , Células Tumorais Cultivadas
2.
Mol Cell Biol ; 13(6): 3577-87, 1993 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8497271

RESUMO

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.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Quinases relacionadas a CDC2 e CDC28 , Ciclo Celular/fisiologia , Quinases Ciclina-Dependentes , Ciclinas/genética , Estradiol/análogos & derivados , Regulação Neoplásica da Expressão Gênica/fisiologia , Substâncias de Crescimento/farmacologia , Mifepristona/farmacologia , Pregnenodionas/farmacologia , Proteínas Serina-Treonina Quinases , Proteína Quinase CDC2/biossíntese , Proteína Quinase CDC2/genética , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Quinase 2 Dependente de Ciclina , Ciclinas/biossíntese , DNA de Neoplasias/análise , DNA de Neoplasias/genética , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Feminino , Fase G1 , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes myc , Histonas/genética , Humanos , Insulina/farmacologia , Cinética , Alcamidas Poli-Insaturadas , Congêneres da Progesterona/farmacologia , Proteínas Quinases/biossíntese , Proteínas Quinases/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes/farmacologia , Fatores de Tempo , Transcrição Gênica/efeitos dos fármacos , Células Tumorais Cultivadas
3.
Oncogene ; 17(26): 3479-91, 1998 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-10030672

RESUMO

Use of the differential display technique to isolate progestin-regulated genes in T-47D human breast cancer cells led to identification of a novel gene, EDD. The cDNA sequence contains a 2799 amino acid open reading frame sharing 40% identity with the predicted 2894 amino acid product of the Drosophila melanogaster tumor suppressor gene hyperplastic discs, while the carboxy-terminal 889 amino acids show 96% identity to a rat 100 kDa HECT domain protein. EDD mRNA was progestin-induced in T-47D cells and was highly abundant in testes and expressed at moderately high levels in other tissues, suggesting a broad role for EDD. Anti-EDD antibodies immunoprecipitated an approximately 300 kDa protein from T-47D cell lysates. HECT family proteins function as E3 ubiquitin-protein ligases, targeting specific proteins for ubiquitin-mediated proteolysis. EDD is likely to function as an E3 as in vitro translated protein bound ubiquitin reversibly through a conserved HECT domain cysteine residue. EDD was localized by FISH to chromosome 8q22, a locus disrupted in a variety of cancers. Given the homology between EDD and the hyperplastic discs protein, which is required for control of imaginal disc growth in Drosophila, EDD potentially has a role in regulation of cell proliferation or differentiation.


Assuntos
Cromossomos Humanos Par 8 , Drosophila melanogaster/genética , Peptídeo Sintases , Proteínas/genética , Proteínas/metabolismo , Sequência de Aminoácidos , Animais , Northern Blotting , Clonagem Molecular , Técnicas Genéticas , Proteínas de Helminto/genética , Humanos , Ligases/genética , Ligases/metabolismo , Masculino , Dados de Sequência Molecular , Pregnenodionas/farmacologia , Ratos , Homologia de Sequência de Aminoácidos , Ubiquitina-Proteína Ligases , Ubiquitinas/metabolismo
4.
Oncogene ; 20(26): 3428-36, 2001 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-11423993

RESUMO

Numerous studies have demonstrated the anticancer activity of the tomato carotenoid, lycopene. However, the molecular mechanism of this action remains unknown. Lycopene inhibition of human breast and endometrial cancer cell growth is associated with inhibition of cell cycle progression at the G(1) phase. In this study we determined the lycopene-mediated changes in the cell cycle machinery. Cells synchronized in the G(1) phase by serum deprivation were treated with lycopene or vehicle and restimulated with 5% serum. Lycopene treatment decreased serum-induced phosphorylation of the retinoblastoma protein and related pocket proteins. This effect was associated with reduced cyclin-dependent kinase (cdk4 and cdk2) activities with no alterations in CDK protein levels. Lycopene caused a decrease in cyclin D1 and D3 levels whereas cyclin E levels did not change. The CDK inhibitor p21(Cip1/Waf1) abundance was reduced while p27(Kip1) levels were unaltered in comparison to control cells. Serum stimulation of control cells resulted in reduction in the p27 content in the cyclin E--cdk2 complex and its accumulation in the cyclin D1--cdk4 complex. This change in distribution was largely prevented by lycopene treatment. These results suggest that lycopene inhibits cell cycle progression via reduction of the cyclin D level and retention of p27 in cyclin E--cdk2, thus leading to inhibition of G(1) CDK activities.


Assuntos
Adenocarcinoma/patologia , Anticarcinógenos/farmacologia , Neoplasias da Mama/patologia , Quinases relacionadas a CDC2 e CDC28 , Carotenoides/farmacologia , Ciclina E/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/metabolismo , Ciclinas/fisiologia , Neoplasias do Endométrio/patologia , Fase G1/efeitos dos fármacos , Proteínas de Neoplasias/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas , Adenocarcinoma/metabolismo , Neoplasias da Mama/metabolismo , Meios de Cultura Livres de Soro/farmacologia , Ciclina D , Quinase 2 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p21 , Neoplasias do Endométrio/metabolismo , Feminino , Sangue Fetal/fisiologia , Humanos , Licopeno , Substâncias Macromoleculares , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteína do Retinoblastoma/metabolismo , Proteína p107 Retinoblastoma-Like , Proteína p130 Retinoblastoma-Like , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
5.
Oncogene ; 8(8): 2127-33, 1993 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8336939

RESUMO

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.


Assuntos
Neoplasias da Mama/genética , Ciclinas/genética , Amplificação de Genes , Regulação Neoplásica da Expressão Gênica , Sequência de Bases , Feminino , Humanos , Dados de Sequência Molecular , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Células Tumorais Cultivadas
6.
Mol Endocrinol ; 11(4): 490-502, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9092801

RESUMO

To define early molecular targets of progestin action, the differential display technique was used to identify genes with altered levels of expression in T-47D breast cancer cells treated with the synthetic progestin ORG 2058 for 3 h. PRG1 was first isolated as a 200-bp cDNA clone and its progestin regulation confirmed by Northern analysis. Cloning of the complete coding region of PRG1 revealed that it shared a high degree of amino acid sequence identity with isoforms of the enzyme 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase from several tissues and species. Expression of PRG1 mRNA was observed in several normal breast epithelial and breast cancer cell lines and in a variety of human tissues, with highest expression in the breast, aorta, and brain. In T-47D cells, PRG1 mRNA was rapidly and transiently induced by progestins, expression peaking between 2 and 4 h and returning to control levels by 12 h. Progestin-induced increases in PRG1 mRNA were inhibited by the progestin antagonist RU 486 and occurred via the progesterone receptor. Progestin induction of PRG1 mRNA was also inhibited by actinomycin D but not by cycloheximide. PRG1 is therefore a novel human gene that is directly regulated by progestins via the progesterone receptor.


Assuntos
Frutose-Bifosfatase/química , Fosfofrutoquinase-1/química , Progestinas/fisiologia , Proteínas/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Neoplasias da Mama/enzimologia , Neoplasias da Mama/genética , Bovinos , Ciclo Celular/genética , Clonagem Molecular , DNA Complementar , Feminino , Frutose-Bifosfatase/genética , Regulação da Expressão Gênica , Glicólise/genética , Humanos , Dados de Sequência Molecular , Fosfofrutoquinase-1/genética , Fosfofrutoquinase-2 , Pregnenodionas/farmacologia , Congêneres da Progesterona/farmacologia , Inibidores da Síntese de Proteínas/farmacologia , Proteínas/química , RNA Mensageiro/isolamento & purificação , Alinhamento de Sequência , Homologia de Sequência do Ácido Nucleico , Transcrição Gênica , Células Tumorais Cultivadas
7.
Mol Endocrinol ; 9(12): 1804-13, 1995 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8614416

RESUMO

To define the mechanisms by which antiestrogens inhibit breast cancer cell proliferation, the effects of the antiestrogen ICI 182780 on G1 cyclins and their cyclin-dependent kinase (CDK) partners were investigated in MCF-7 cells. Inhibition of entry into S phase became evident 9 h after treatment, with the proportion of cells in S phase reaching a minimum by 24 h. ICI 182780 increased the proportion of the hypophosphorylated, growth inhibitory form of the retinoblastoma protein (pRB). This change began at 4-6 h, preceding effects on S phase. This suggests that there are early effects on the activities of CDKs that target pRB that are not merely a consequence of changes in cell cycle progression. The kinase activity of Cdk2 decreased to low levels at 18-24 h when changes in S phase and pRB phosphorylation were well advanced. An earlier effect was seen on kinase activity associated with immunoprecipitated cyclin D1, which was reduced approximately 40% by 12 h, with further decreases at 18-24 h. Cdk2 and Cdk4 protein levels remained constant over 24 h. Cyclin D1 messenger RNA and protein were down-regulated by ICI 182780 from 2 h, with levels halved at 8 h. ICI 182780 also increased the expression of the CDK inhibitors p27KIP1 and p21WAF1/CIP1 at later times. These observations are compatible with the hypothesis that antiestrogens block entry of cells into S phase and inhibit cell proliferation as the consequence of an early decline in pRB phosphorylation contributed to by reduced cyclin D1/Cdk4 activity. At later times, increased CDK inhibitor abundance may act to repress Cdk2 and Cdk4 activities, causing additional reductions in pRB phosphorylation, thus maintaining the antiestrogen blockade of cell cycle progression.


Assuntos
Neoplasias da Mama/patologia , Quinases relacionadas a CDC2 e CDC28 , Ciclo Celular/efeitos dos fármacos , Quinases Ciclina-Dependentes/antagonistas & inibidores , Antagonistas de Estrogênios/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas , Proteína do Retinoblastoma/metabolismo , Ciclina D1 , Quinase 2 Dependente de Ciclina , Quinase 4 Dependente de Ciclina , Ciclinas/genética , Ciclinas/metabolismo , Estradiol/análogos & derivados , Estradiol/farmacologia , Fulvestranto , Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Fosforilação , Células Tumorais Cultivadas
8.
J Bone Miner Res ; 9(8): 1251-8, 1994 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-7976507

RESUMO

Estrogenic effects on the proliferation and differentiated cellular functions of bone cells have been described in vivo and in vitro. In particular, stimulatory effects on the growth rate of osteoblasts have been observed, although these are generally small. In an attempt to produce a more sensitive model for the study of estrogen action in bone, HTB 96 human osteoblast-like osteosarcoma cells, which lack endogenous estrogen receptor (ER), were stably transfected with an expression vector coding for the human ER gene. Several HTB 96 sublines expressing ER protein, detected by ligand binding and immunoassay, were isolated. The ability of 17 beta-estradiol (E2) to induce chloramphenicol acetyltransferase (CAT) activity from a cotransfected reporter vector containing the CAT gene linked to the Xenopus vitellogenin A2 gene estrogen response element demonstrated that the expressed ER was functional. ER continued to be expressed over a 30 week culture period. E2 but not other steroids significantly reduced growth rates and produced an altered morphology in HTB 96 sublines expressing higher levels of ER. The antiestrogen 4-hydroxytamoxifen partially reversed the E2 effect on growth rate. Transient transfection of cells expressing ER with a vector containing the CAT gene linked to the mouse mammary tumor virus long terminal repeat sequence, which contains response elements for the glucocorticoid receptor but not the ER, showed that E2 was able to inhibit CAT induction by dexamethasone. This result suggest that in ER-transfected HTB 9 cells the effects of E2 may result not from direct activation of endogenous genes but instead by transcriptional interference.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Estradiol/farmacologia , Regulação da Expressão Gênica , Osteossarcoma/genética , Receptores de Estrogênio/genética , Tamoxifeno/análogos & derivados , Animais , Sequência de Bases , Divisão Celular/efeitos dos fármacos , Cloranfenicol O-Acetiltransferase/genética , Cloranfenicol O-Acetiltransferase/metabolismo , Genes Reporter/fisiologia , Vetores Genéticos , Humanos , Dados de Sequência Molecular , Osteossarcoma/fisiopatologia , Receptores de Estrogênio/biossíntese , Receptores de Estrogênio/fisiologia , Tamoxifeno/farmacologia , Transfecção , Xenopus laevis
9.
J Med Chem ; 29(12): 2511-9, 1986 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-3783610

RESUMO

A series of triarylethylene compounds related to 4-hydroxyclomiphene (2) in which the vinyl Cl substituent was replaced by ethyl (5), Br (6), H (7), CN (8), or NO2 (9) substituents were synthesized to facilitate studies of the molecular actions of synthetic nonsteroidal antiestrogens. The relative binding affinities of these compounds for the estrogen receptor (ER) and the antiestrogen binding site (AEBS) in MCF 7 human mammary carcinoma cells were measured and correlated with the effects of these drugs on cell proliferation kinetics. Affinities for ER and AEBS were highly correlated, illustrating that vinyl substituents influence binding to ER and AEBS in a parallel manner. All compounds except 7 had biphasic effects on cell proliferation kinetics, indicating the presence of at least two distinct mechanisms by which hydroxytriarylethylenes inhibit breast cancer cell proliferation. In the concentration range 10(-10)-10(-8) M, cell proliferation was inhibited by 60-70%, these effects were estrogen-reversible, and the degree of growth inhibition was in the order Cl greater than Et greater than Br greater than NO2 greater than CN greater than H, which paralleled the order of affinities for ER. There was no further inhibition of cell growth between 10(-8) and 10(-6) M, but at concentrations greater than 10(-6) M there was a further dose-dependent decrease in cell growth mediated by mechanisms yet to be defined but apparently distinct from ER-mediated events. In both concentration ranges, growth inhibition was accompanied by accumulation of cells in the G1 phase of the cell cycle. These data, obtained with a novel series of hydroxytriarylethylenes, have enabled clear definition of two distinct mechanisms of growth inhibition by triarylethylene antiestrogens. They also indicate that among the vinyl substitutions examined to date the Cl substituent yields the most active molecule both in terms of affinity for ER and AEBS and potency as a growth inhibitory agent.


Assuntos
Antineoplásicos/síntese química , Estilbenos/síntese química , Neoplasias da Mama/patologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Feminino , Humanos , Indicadores e Reagentes , Espectroscopia de Ressonância Magnética , Estilbenos/toxicidade , Relação Estrutura-Atividade
10.
J Steroid Biochem Mol Biol ; 47(1-6): 99-106, 1993 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8274447

RESUMO

Recent advances in defining the molecular mechanisms of cell cycle control in eukaryotes provide a basis for better understanding the hormonal control of cell proliferation in normal and neoplastic breast epithelium. It is now clear that a number of critical steps in cell cycle progression are controlled by families of serine/threonine kinases, the cdks. These kinases are activated by interactions with various cyclin gene products which form the regulatory subunits of the kinase complexes. Several families of cyclins control cell cycle progression in G1 phase, cyclins C, D and E, or in S, G2 and mitosis, cyclins A and B. Recent studies have defined the expression and regulation of cyclin genes in normal breast epithelial cells and in breast cancer cell lines. Following growth arrest of T-47D breast cancer cells by serum deprivation restimulation with insulin results in sequential induction of cyclin genes. Cyclin D1 mRNA increases within 1 h of mitogenic stimulation and is followed by increased expression of cyclins D3 and E in G1 phase, cyclin A in late G1/early S phase and cyclin B1 in G2. Similar results were observed following epidermal growth factor stimulation of normal breast epithelial cells. Other hormones--oestrogens and progestins--and growth factors--insulin-like growth factor-I and basic fibroblast growth factor--with actions in G1 were also investigated for their effects on G1 cyclin gene expression. In all cases there was an excellent correlation between the induction of cyclin D1 mRNA and subsequent entry into S phase. Furthermore, growth inhibition by antioestrogens and concurrent G1 arrest were preceded by an acute decrease in cyclin D1 gene expression. These observations suggest a likely role for cyclin D1 in mediating many of the known hormonal effects on cell proliferation in breast epithelial cells.


Assuntos
Neoplasias da Mama/patologia , Mama/patologia , Ciclo Celular/fisiologia , Ciclinas/genética , Expressão Gênica , Divisão Celular/efeitos dos fármacos , Ciclinas/farmacologia , Epitélio/patologia , Humanos , RNA Mensageiro/metabolismo
11.
J Steroid Biochem Mol Biol ; 41(3-8): 529-36, 1992 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-1562523

RESUMO

The mechanisms underlying loss of oestrogen responsiveness in breast cancer are not well-defined. Potential mechanisms include loss of receptor expression, alterations in the oestrogen receptor (ER) gene producing proteins with abnormal function, or changes to receptor-dependent or -independent pathways controlling cell proliferation. Examination by Southern analysis of the ER gene in a series of ER-negative and -positive breast tumour biopsies failed to provide evidence of gross rearrangements and in only one of thirty seven tumour DNA samples was significant gene amplification observed. No restriction fragment length polymorphisms were detected for the restriction enzymes EcoR I, Pst I or Hind III. Methylation of the ER gene as assessed by Hpa II and Msp I restriction enzyme digests varied between tumours but the degree of methylation was not correlated with levels of expression of the receptor protein. Similar findings applied in a series of ER-negative and -positive breast cancer cell lines and clonal lines of MCF-7 cells, which were developed as an in vitro model for the acquisition of oestrogen and antioestrogen resistance. In this model there was no evidence that changes to ER receptor function and/or structure at the level of the ER gene, mRNA, ligand binding, and ability to induce progesterone receptor might account for the development of hormone resistance. However, the ability of ER to interact with a DNA sequence containing the vitellogenin promoter oestrogen response element, as assessed by gel retardation assay, was impaired in the clone showing the greatest degree of oestrogen and antioestrogen resistance.


Assuntos
Neoplasias da Mama/genética , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Sequência de Bases , Neoplasias da Mama/metabolismo , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Células Clonais , DNA de Neoplasias/genética , DNA de Neoplasias/isolamento & purificação , Estradiol/farmacologia , Feminino , Humanos , Dados de Sequência Molecular , Receptores de Progesterona/metabolismo , Sequências Reguladoras de Ácido Nucleico , Mapeamento por Restrição , Tamoxifeno/farmacologia , Células Tumorais Cultivadas
12.
Phys Sportsmed ; 14(3): 99-104, 1986 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27467344

RESUMO

In brief: To determine what public and individual efforts might reduce bicycling accidents and injuries, a survey was conducted during the primary cycling months (April through September) in Boulder, Colorado. Of 253 patients (87 women, 166 men, average age 22) treated for injuries in bicycling accidents, almost 30% of the accidents were caused by gravel, and nearly half involved a motor vehicle. The most frequent types of injury were abrasions, contusions, lacerations, and fractures. The survey results confirm the need for adequate head protection, satisfactory lighting, and increased awareness on the bicyclist's part. In addition, improved street maintenance will drastically reduce the number of cycling injuries and accidents.

14.
Br J Cancer ; 98(6): 1085-93, 2008 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-18349819

RESUMO

Despite a high initial response rate to first-line platinum/paclitaxel chemotherapy, most women with epithelial ovarian cancer relapse with recurrent disease that becomes refractory to further cytotoxic treatment. We have previously shown that the E3 ubiquitin ligase, EDD, a regulator of DNA damage responses, is amplified and overexpressed in serous ovarian carcinoma. Given that DNA damage pathways are linked to platinum resistance, the aim of this study was to determine if EDD expression was associated with disease recurrence and platinum sensitivity in serous ovarian cancer. High nuclear EDD expression, as determined by immunohistochemistry in a cohort of 151 women with serous ovarian carcinoma, was associated with an approximately two-fold increased risk of disease recurrence and death in patients who initially responded to first-line chemotherapy, independently of disease stage and suboptimal debulking. Although EDD expression was not directly correlated with relative cisplatin sensitivity of ovarian cancer cell lines, sensitivity to cisplatin was partially restored in platinum-resistant A2780-cp70 ovarian cancer cells following siRNA-mediated knockdown of EDD expression. These results identify EDD as a new independent prognostic marker for outcome in serous ovarian cancer, and suggest that pathways involving EDD, including DNA damage responses, may represent new therapeutic targets for chemoresistant ovarian cancer.


Assuntos
Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos , Neoplasias Ovarianas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Linhagem Celular Tumoral , Cistadenocarcinoma Seroso , Feminino , Humanos , Pessoa de Meia-Idade , Recidiva Local de Neoplasia , Neoplasias Ovarianas/tratamento farmacológico , Prognóstico , Estudos Retrospectivos
15.
Mol Pharmacol ; 31(5): 541-51, 1987 May.
Artigo em Inglês | MEDLINE | ID: mdl-3553893

RESUMO

Synthetic nonsteroidal antiestrogens are bound intracellularly by two high affinity saturable bindings sites, the estrogen receptor and the microsomal antiestrogen-binding site (AEBS). In order to further define the structural requirements for ligand binding to AEBS from rat liver and the MCF 7 human breast cancer cell line, the relative binding affinities of an extensive series of structurally related ligands were investigated using competitive binding assay techniques. The groups of compounds studied were: analogues of the triphenylethylene antiestrogens, Cl 628 and tamoxifen; analogues of cyclofenil; bibenzyl and stilbene derivatives; analogues of the cytochrome P-450 inhibitor SKF-525A; phenothiazine derivatives; and a series of structurally related compounds with a variety of pharmacological activities. High affinity binding to AEBS required the presence of both a hydrophilic basic aminoether side chain and a hydrophobic aromatic ring structure (di- or tricyclic for maximal affinity). Structural modifications to either influenced binding affinity. Aromatic substitution either raised (CF3) or lowered (OH, OCH3) affinity, apparently by electronic effects transmitted through the benzene nucleus. Side chain structure was the major determinant of binding affinity, but its influence was complex and dependent upon terminal amino group structure, side chain branching and substitution, and tissue source of AEBS. Optimal binding affinity was shown by side chains bearing basic heterocyclic amino terminal groups. Other cellular sites that are known to bind antiestrogens with relatively high affinity include calmodulin, cytochrome P-450, and histamine, dopamine, and muscarinic receptors. Binding studies using a variety of pharmacologically active and radiolabeled ligands selective for these sites, including those for dopamine D1 and D2 receptors ([3H]fluphenazine, [3H]flupenthixol, [3H]spiperone, and [3H]SCH 23390) and histamine H1 receptors ([3H]pyrilamine), demonstrated that several of these compounds interact with AEBS with high affinity. However, the ligand specificity and other binding properties of the AEBS as determined by competitive binding studies and Scatchard analysis show this site to be a molecular entity truly distinct from these other cellular binding sites.


Assuntos
Antagonistas de Estrogênios/metabolismo , Microssomos/metabolismo , Receptores de Droga , Receptores de Estrogênio/metabolismo , Animais , Neoplasias da Mama/metabolismo , Linhagem Celular , Ciclofenil/análogos & derivados , Ciclofenil/metabolismo , Humanos , Nitromifeno/análogos & derivados , Nitromifeno/metabolismo , Fenotiazinas/metabolismo , Proadifeno/análogos & derivados , Proadifeno/metabolismo , Ratos , Estilbenos/metabolismo , Relação Estrutura-Atividade , Tamoxifeno/análogos & derivados , Tamoxifeno/metabolismo , Xantinas/metabolismo
16.
Biochem J ; 236(3): 903-11, 1986 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-3790095

RESUMO

The properties of an antioestrogen binding site (AEBS), which has high affinity and specificity for nonsteroidal antioestrogens and structurally related compounds, have been studied in rat liver microsomes. When subcellular organelles were separated on Percoll density gradients the distribution of the AEBS paralleled that of NADPH-cytochrome c reductase, indicating that the AEBS is associated with the endoplasmic reticulum. Saturation analysis showed that [3H]tamoxifen was bound to a single class of saturable binding sites in liver microsomes with a KD of 0.9 +/- 0.1 nM at 0 degrees C. The equilibrium KD was not significantly different at 22 degrees C. The KD calculated from the association and dissociation rate constants for [3H]tamoxifen binding at 0 degrees C and 22 degrees C was compatible with the KD measured at equilibrium. Ligand specificity studies using tamoxifen analogues showed qualitatively similar structure-affinity relationships for the AEBS from both rat liver and the MCF 7 breast cancer cell line. In general structural modifications caused correspondingly greater changes in affinity for rat liver AEBS than for MCF 7 AEBS. The AEBS was solubilized from microsomal membranes with sodium cholate. This was the only detergent of nine tested that solubilized the site in high yield without loss of activity. Solubilization using cholate was more effective in the presence of 1 M-NaCl. In the solubilized state there was an apparent loss of [3H]tamoxifen binding activity which could be restored by dilution of the detergent. Gel filtration indicated an Mr of 440,000-490,000 for the AEBS-cholate complex. These studies demonstrate that rat liver contains high concentrations of a microsomal AEBS which has similar properties and specificity to the AEBS previously described in human breast cancer cells. This site can be solubilized by sodium cholate to supply material suitable for further purification.


Assuntos
Antagonistas de Estrogênios/metabolismo , Microssomos Hepáticos/metabolismo , Receptores de Droga , Receptores de Estrogênio/metabolismo , Animais , Sítios de Ligação , Ácido Cólico , Ácidos Cólicos/farmacologia , Cromatografia em Gel , Detergentes/farmacologia , Feminino , Ligantes , Masculino , Microssomos Hepáticos/efeitos dos fármacos , Ratos , Ratos Endogâmicos , Frações Subcelulares/metabolismo , Tamoxifeno/análogos & derivados , Tamoxifeno/metabolismo
17.
Biochem Biophys Res Commun ; 120(1): 109-15, 1984 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-6712683

RESUMO

Saturation and competitive binding analyses demonstrated the presence of a high affinity (KD = 0.92 nM), specific antiestrogen binding site (AEBS) in rat liver microsomes and at least 75% of total liver AEBS was recovered in this fraction. When microsomes were further separated into smooth and rough fractions, AEBS was concentrated in the latter. Subsequent dissociation of ribosomes from the rough membranes revealed that AEBS was associated with the membrane and not the ribosomal fraction. Antiestrogen binding activity could not be extracted from membranes with 1 M KCl or 0.5 M acetic acid but could be solubilized with sodium cholate. These data indicate that AEBS is an integral membrane component of the rough microsomal fraction of rat liver.


Assuntos
Membranas Intracelulares/metabolismo , Microssomos Hepáticos/metabolismo , Receptores de Droga , Receptores de Estrogênio/metabolismo , Animais , Ligação Competitiva , Feminino , Fígado/metabolismo , Ratos , Ratos Endogâmicos , Receptores de Estrogênio/isolamento & purificação , Solubilidade , Frações Subcelulares/metabolismo
18.
J Steroid Biochem ; 34(1-6): 483-90, 1989.
Artigo em Inglês | MEDLINE | ID: mdl-2626043

RESUMO

The oestrogen receptor (ER) gene was introduced into an ER-negative osteoblast-like osteosarcoma cell line HTB 96 by transfection. A number of clones were isolated which expressed ER at levels of up to 70 fmol/mg cytosol protein as determined by immunoassay. Scatchard analysis of the binding of [3H]17 beta-oestradiol in cytosols demonstrated the presence of high affinity binding sites, with a dissociation constant of 0.08-0.13 nM at 4 degrees C. High levels of a 3 kb ER mRNA are produced by the clones, which have gene copy numbers ranging from 2 to greater than 10. Functional receptor activity has been demonstrated by co-transfection of a plasmid containing the chloramphenicol acetyl transferase (CAT) gene linked to an oestrogen response element. Induction of CAT activity is observed in the presence of added oestradiol and is concentration-dependent. The transfected ER is also able to affect endogenous cellular function as several ER-positive clones, but not HTB 96 cells, are growth inhibited by oestradiol in the concentration range 10(-9)-10(-7) M. These effects on growth are not induced by other classes of steroids and are reversible by antioestrogens. No endogenous genes have yet been identified which are oestrogen-regulated in ER-transfected clones.


Assuntos
Receptores de Estrogênio/genética , Transfecção , Células Tumorais Cultivadas/metabolismo , Sequência de Bases , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Estradiol/farmacologia , Vetores Genéticos , Humanos , Cinética , Dados de Sequência Molecular , Osteoblastos/metabolismo , Osteossarcoma , Plasmídeos , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/efeitos dos fármacos
19.
J Biol Chem ; 259(7): 4223-9, 1984 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-6707003

RESUMO

In the presence of estradiol, at a concentration sufficient to saturate the estrogen receptor, the antiestrogenic and anti-tumor agent tamoxifen was bound to a high affinity (KD = 0.97 +/- 0.15 nM at 4 degrees C) saturable binding site (141,300 +/- 20,100 sites/cell) in MCF 7 human mammary carcinoma cells. The distribution of this site between cytosol, mitochondrial, microsomal (heavy and light), and nuclear fractions paralleled that of NADPH-cytochrome c reductase, an enzyme marker for endoplasmic reticulum. The interaction between tamoxifen and the high affinity site was influenced by changes in pH, ionic strength, and temperature. The kinetic rate constants k+1 and k-1 showed strong temperature dependence, but KD was unaffected by changes in temperature. Competition studies employing analogs of the anit-estrogens tamoxifen, clomiphene, and CI 628 revealed narrow specificity for triphenylethylene derivatives with basic ether side chains.


Assuntos
Neoplasias da Mama/metabolismo , Antagonistas de Estrogênios/metabolismo , Microssomos/metabolismo , Receptores de Droga , Receptores de Estrogênio/metabolismo , Tamoxifeno/análogos & derivados , Tamoxifeno/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Citosol/metabolismo , Feminino , Humanos , Cinética , Mitocôndrias/metabolismo , Relação Estrutura-Atividade
20.
Biochem Biophys Res Commun ; 140(2): 523-9, 1986 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-3778464

RESUMO

Treatment of MCF 7 human breast cancer cells with three high affinity hydroxylated antiestrogens (Kd for the estrogen receptor = 0.11-0.45 nM) resulted in biphasic inhibition of cell growth. Administration of 0.1-1.0 nM of each drug caused a concentration-dependent decrease in cell number to a maximum of 30-40% of control but no further change was observed as the drug concentration was increased to 1 microM. Between 1.0 and 10 microM, however, a further concentration-dependent decrease in cell proliferation was observed. Among these compounds relative potencies paralleled their affinities for estrogen receptor in the 0.1-10 nM range but at micromolar concentrations this relationship did not hold. It is concluded that antiestrogens inhibit cell proliferation by two distinct mechanisms one of which involves the estrogen receptor and the other a mechanism yet to be defined. The parallel changes in cell cycle kinetic parameters accompanying growth inhibition in both concentration ranges i.e. accumulation of cells in the G1 phase at the expense of S phase cells, suggests that both mechanisms may converge on common pathways critical to cell cycle progression.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Clomifeno/análogos & derivados , Antagonistas de Estrogênios/farmacologia , Receptores de Estrogênio/efeitos dos fármacos , Ligação Competitiva , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Clomifeno/farmacologia , Estradiol/farmacologia , Antagonistas de Estrogênios/metabolismo , Humanos , Estilbenos/farmacologia , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA