Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
1.
Br J Cancer ; 95(9): 1220-8, 2006 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-17043687

RESUMO

Activation of the type I insulin-like growth factor receptor (IGF-IR) regulates several aspects of the malignant phenotype, including cancer cell proliferation and metastasis. Phosphorylation of adaptor proteins downstream of IGF-IR may couple IGF action to specific cancer phenotypes. In this study, we sought to determine if insulin receptor substrate-1 and -2 (IRS-1 and -2) mediate distinct biological effects in breast cancer cells. Insulin receptor substrate-1 and IRS-2 were expressed in T47D-YA breast cancer cells, which lack IRS-1 and -2 expression, yet retain functional IGF-IR. In the absence of IRS-1 and -2 expression, IGF-IR activation was unable to stimulate proliferation or motility in T47D-YA cells. Expression of IRS-1 resulted in IGF-I-stimulated proliferation, but did not affect motility. In contrast, expression of IRS-2 enhanced IGF-I-stimulated motility, but did not stimulate proliferation. The alphaIR-3, an inhibitor of the IGF-IR, was unable to affect these IGF-stimulated phenotypes unless IRS-1 or -2 was expressed. Thus, IGF-IR alone is unable to regulate important breast cancer cell phenotypes. In these cells, IRS proteins are required for and mediate distinct aspects of IGF-IR-stimulated behaviour. As multiple agents targeting the IGF-IR are currently in early clinical trials, IRS expression should be considered as a potential biomarker for IGF-IR responsiveness.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Fosfoproteínas/fisiologia , Receptor IGF Tipo 1/fisiologia , Anticorpos Monoclonais/farmacologia , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Neoplasias da Mama/fisiopatologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Substratos do Receptor de Insulina , Fator de Crescimento Insulin-Like I/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Fosfoproteínas/genética , Interferência de RNA , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Transfecção
2.
Breast Cancer Res Treat ; 94(2): 171-83, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16175315

RESUMO

Progesterone in hormone replacement therapy (HRT) preparations increases, while hysterectomy greatly reduces, the incidence of breast cancer. Cross-talk between the progesterone and growth factor signaling pathways occurs at multiple levels and this maybe a key factor in breast cancer survival and progression. To test this hypothesis, we characterized the effect of progesterone pre-treatment on the sensitization of the epidermal growth factor (EGF) signaling pathway to EGF in the breast cancer cell line ZR-75. For the first time in ZR-75 cells and in agreement with previous work using synthetic progestins, we demonstrate that pre-treatment with the natural ligand progesterone increases EGF receptor (EGFR) levels and subsequent ligand-dependent phosphorylation. Downstream we demonstrate that progesterone alone increases erk-1 + 2 phosphorylation, potentiates EGF-phosphorylated erk-1 + 2 and maintains these levels elevated for 24 h; over 20 h longer than in vehicle treated cells. Additionally, progesterone increased the levels of STAT5, another component of the EGF signaling cascade. Progesterone increased EGF mediated transcription of a c-fos promoter reporter and the nuclear localization of the native c-fos protein. Furthermore, progesterone and EGF both alone and in combination, significantly increase cell proliferation. Several results presented herein demonstrate the conformity between the action of the natural ligand progesterone with that of synthetic progestins such as MPA and R5020 and allows the postulation that the progestin/progesterone-dependent increase of EGF signaling provides a survival advantage to burgeoning cancer cells and may contribute to the breast cancer risk associated with endogenous progesterone and with progestin-containing HRT.


Assuntos
Neoplasias da Mama/metabolismo , Fator de Crescimento Epidérmico/efeitos dos fármacos , Receptores ErbB/metabolismo , Progesterona/farmacologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Fosforilação/efeitos dos fármacos , Progesterona/administração & dosagem , Transdução de Sinais/efeitos dos fármacos
3.
J Biol Chem ; 276(43): 39843-51, 2001 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-11546784

RESUMO

Progesterone receptors (PR) contain three activation functions (AFs) that together define the extent to which they regulate transcription. AF1 and AF2 are common to the two isoforms of PR, PR-A and PR-B, whereas AF3 lies within the N-terminal 164 amino acids unique to PR-B, termed the "B-upstream segment" (BUS). To define the BUS regions that contribute to AF3 function, we generated a series of deletion and amino acid substitution mutants and tested them in three backgrounds as follows: BUS alone fused to the PR DNA binding domain (BUS-DBD), the entire PR-B N terminus linked to its DBD (NT-B), and full-length PR-B. Analyses of these mutants identified two regions in BUS whose loss reduces AF3 activity by more than 90%. These are associated with amino acids 54-90 (R1) and 120-154 (R2). R1 contains a consensus (55)LXXLL(59) motif (L1) identical to ones found in nuclear receptor co-activators. R2 is adjacent to a second nuclear receptor box (L2) at (115)LXXLL(119) and contains a conserved tryptophan (Trp-140). Their mutation completely disrupts AF3 activity in a promoter and cell type-independent manner. Critical mutations elicited similar effects on all three B-receptor backgrounds. This underscores the probability that these mutations alter a process linking BUS structure to the function of full-length PR-B in a fundamental way.


Assuntos
Receptores de Progesterona/metabolismo , Proteínas de Saccharomyces cerevisiae , Fatores de Transcrição/metabolismo , Motivos de Aminoácidos , Sítios de Ligação , Humanos , Proteínas de Membrana , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fragmentos de Peptídeos , Deleção de Sequência , Transcrição Gênica
4.
Mol Cell Biol ; 21(18): 6122-31, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11509655

RESUMO

Breast cancers often exhibit elevated expression of tyrosine kinase growth factor receptors; these pathways influence breast cancer cell growth in part by targeting steroid hormone receptors, including progesterone receptors (PR). To mimic activation of molecules downstream of growth factor-initiated signaling pathways, we overexpressed mitogen-activated protein kinase (MAPK; also known as extracellular signal-regulated kinase) kinase kinase 1 (MEKK1) in T47D human breast cancer cells expressing the B isoform of PR. MEKK1 is a strong activator of p42 and p44 MAPKs. MEKK1 expression increased progestin-mediated transcription 8- to 10-fold above normal PR-driven transcription levels. This was dependent on the presence of a progesterone response element and functional PR. PR protein levels were unchanged by MEKK1 alone but were extensively down-regulated by MEKK1 plus the progestin R5020. MEKK1 expression resulted in phosphorylation of PR on Ser294, a MAPK consensus site known to mediate ligand-dependent PR degradation. MEK inhibitors blocked phosphorylation of Ser294 and attenuated PR transcriptional hyperactivity in response to MEKK1 plus R5020; stabilization of PR by inhibition of the 26S proteasome produced similar results. T47D cells stably expressing mutant S294A PR, in which serine 294 is replaced by alanine, fail to undergo ligand-dependent down-regulation and are resistant to MEKK1-plus-R5020-induced transcriptional synergy but respond to progestins alone. Similarly, c-myc protein levels are synergistically increased by epidermal growth factor and R5020 in cells expressing wild-type PR, but not S294A PR. Thus, highly stable mutant PR are functional in response to progestins but are incapable of cross talk with MAPK-driven pathways. These studies demonstrate a paradoxical coupling between steroid receptor down-regulation and transcriptional hyperactivity. They also suggest a link between phosphorylation of PR by MAPKs in response to peptide growth factor signaling and steroid hormone control of breast cancer cell growth.


Assuntos
MAP Quinase Quinase Quinase 1 , Proteínas Serina-Treonina Quinases/fisiologia , Receptores de Progesterona/fisiologia , Transdução de Sinais , Regulação para Baixo , Feminino , Humanos , Ligantes , Sistema de Sinalização das MAP Quinases/fisiologia , Fosforilação , Serina , Ativação Transcricional , Células Tumorais Cultivadas
5.
J Biol Chem ; 276(26): 23825-31, 2001 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-11328821

RESUMO

To understand the basis for functional differences between the two human progesterone receptors (PR), we have carried out a detailed biochemical and biophysical analysis of the N-terminal region of each isoform. Extending our previous work on the A-isoform (Bain, D. L, Franden, M. A., McManaman, J. L., Takimoto, G. S., and Horwitz, K. B. (2000) J. Biol. Chem. 275, 7313-7320), here we present studies on the N-terminal region of the B-isoform (NT-B) and compare its properties to its A-receptor counterpart (NT-A). As seen previously with NT-A, NT-B is quantitatively monomeric in solution, yet undergoes N-terminal-mediated assembly upon DNA binding. Limited proteolysis, microsequencing, and sedimentation analyses indicate that the B-isoform exists in a non-globular, extended conformation very similar to that of NT-A. Additionally, the 164 amino acids unique to the B-isoform (BUS) appear to be in a more extended conformation relative to sequences common to both receptors and do not exist as an independent structural domain. However, sedimentation studies of NT-A and NT-B show differences in the ensemble distribution of their conformational states. We hypothesize that isoform-specific functional differences are not due to structural differences, per se. Rather, the transcriptional element BUS, or possibly other transcription factors, causes a redistribution of the conformational ensemble by stabilizing a more functionally active set of conformations in NT-B.


Assuntos
Receptores de Progesterona/química , Humanos , Cinética , Conformação Proteica , Receptores de Progesterona/metabolismo , Elementos de Resposta , Transcrição Gênica , Ultracentrifugação
6.
Breast Cancer Res Treat ; 69(1): 69-79, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11759830

RESUMO

A major goal of cancer chemotherapy is the identification of cytotoxic compounds that are highly selective for cancer cells. We describe here one such compound - a novel iron chelator, desferri-exochelin 772SM. This desferri-exochelin has unique chemical and pharmacological properties, including extremely high iron binding affinity, the capacity to block iron-mediated redox reactions, and lipid solubility which enables it to enter cells rapidly. At low concentrations, this desferri-exochelin kills T47D-YB and MCF-7 human breast cancer cells by inducing apoptosis, but only reversibly arrests the growth of normal human mammary epithelial cells without cytotoxicity. Since iron-loaded exochelin is ineffective, iron chelation accounts for the efficacy of desferri-exochelin. For both the killing of breast cancer cells and the growth arrest of normal breast epithelial cells, desferri-exochelin was effective at much lower concentrations than the lipid-insoluble iron chelator deferoxamine, which has shown only limited potential as an anti-cancer agent. Growth arrest of progesterone receptor positive T47D-YB cells with the progestin R5020 transiently protects them from the cytotoxic effects of desferri-exochelin, but the cells are killed after cell growth resumes. Similarly, MCF-7 cells arrested with the estrogen antagonist ICI182780 are transiently resistant to killing by desferri-exochelin. Thus the desferri-exochelin is cytotoxic only to actively growing tumor cells. Since desferri-exochelin 772SM can selectively and efficiently destroy proliferating cancer cells without damaging normal cells, it may prove useful for the treatment of cancer.


Assuntos
Apoptose , Neoplasias da Mama/patologia , Mama/citologia , Quelantes de Ferro/farmacologia , Peptídeos Cíclicos/farmacologia , Divisão Celular , Relação Dose-Resposta a Droga , Feminino , Humanos , Células Tumorais Cultivadas
7.
Steroids ; 65(10-11): 579-84, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11108862

RESUMO

The development of tamoxifen resistance and consequent disease progression are common occurrences in breast cancers, often despite the continuing expression of estrogen receptors (ER). Tamoxifen is a mixed antagonist, having both agonist and antagonist properties. We have suggested that the development of tamoxifen resistance is associated with an increase in its agonist-like properties, resulting in loss of antagonist effects or even inappropriate tumor stimulation. Nuclear receptor function is influenced by a family of transcriptional coregulators, that either enhance or suppress transcriptional activity. Using a mixed antagonist-biased two-hybrid screening strategy, we identified two such proteins: the human homolog of the nuclear receptor corepressor, N-CoR, and a novel coactivator, L7/SPA (Switch Protein for Antagonists). In transcriptional studies, N-CoR suppressed the agonist properties of tamoxifen and RU486, and L7/SPA increased agonist effects. We speculated that the relative levels of these coactivators and corepressors may determine the balance of agonist and antagonist properties of mixed antagonists, such as tamoxifen. Using quantitative RT-PCR, we, therefore, measured the levels of transcripts encoding these coregulators, as well as the corepressor SMRT, and the coactivator SRC-1, in a small cohort of tamoxifen-resistant and sensitive breast tumors. The results suggest that tumor sensitivity to mixed antagonists may be governed by a complex set of transcription factors, which we are only now beginning to understand.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Receptores de Estrogênio/genética , Tamoxifeno/farmacologia , Neoplasias da Mama/patologia , Resistência a Medicamentos , Feminino , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/farmacologia , Correpressor 1 de Receptor Nuclear , Receptores de Estrogênio/química , Proteínas Repressoras/genética , Proteínas Repressoras/farmacologia , Fatores de Transcrição/genética , Fatores de Transcrição/farmacologia
8.
Circulation ; 101(15): 1792-8, 2000 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-10769279

RESUMO

BACKGROUND: Estrogens have vascular effects through the activation of estrogen receptors (ERs). In addition to ERalpha, the first ER to be cloned, a second subtype called ERbeta has recently been discovered. METHODS AND RESULTS: Using a reverse-transcriptase polymerase chain reaction assay that employs the same primer pair to simultaneously amplify ERalpha and ERbeta transcripts, we found that ERbeta is the ER form that is predominantly expressed in human vascular smooth muscle, particularly in women. The transcriptional effects of the 2 ERs in transfected HeLa cells differed. In response to 17beta-estradiol, ERalpha is a stronger transactivator than ERbeta at low receptor concentrations. However, at higher receptor concentrations, ERalpha activity self-squelches, and ERbeta is a stronger transactivator. Tamoxifen has partial agonist effects with ERalpha but not with ERbeta. CONCLUSIONS: The protective effects of estrogens in the cardiovascular system of women may be due to the genomic effects of ERbeta in vascular tissue.


Assuntos
Músculo Liso Vascular/metabolismo , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Transcrição Gênica , Estradiol/análogos & derivados , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Feminino , Fulvestranto , Células HeLa , Humanos , Ligantes , Regiões Promotoras Genéticas , RNA/análise , Receptores de Estrogênio/efeitos dos fármacos , Análise de Regressão , Tamoxifeno/farmacologia
9.
J Mol Endocrinol ; 24(2): 165-82, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10750018

RESUMO

Ligand-activated progesterone receptors (PR) bind to DNA at specific progesterone response elements by means of a DNA binding domain (DBD(PR)) containing two highly conserved zinc fingers. DNA-bound PRs regulate transcription via interaction with other nuclear proteins and transcription factors. We have now identified four HeLa cell nuclear proteins that copurify with a glutathionine-S-transferase-human DBD(PR )fusion protein. Microsequence and immunoblot analyses identified one of these proteins as the 113 kDa poly(ADP-ribose) polymerase. The three other proteins were identified as subunits of the DNA-dependent protein kinase (DNA-PK) holoenzyme: its DNA binding regulatory heterodimers consisting of Ku70 and Ku86, and the 460 kDa catalytic subunit, DNA-PK(CS). DNA-PK that was 'pulled-down' by DBD(PR) on the affinity resin was able to (1) autophosphorylate Ku70, Ku86, and DNA-PK(CS), (2) transphosphorylate DBD(PR), and (3) phosphorylate a DNA-PK-specific p53 peptide substrate. DNA-PK was also able to associate with the DBD of the yeast activator GAL4. However, neither a PR DBD mutant lacking a structured first zinc finger (DBD(CYS)) nor the core DBD of the estrogen receptor (DBD(ER)) copurified DNA-PK, suggesting the interaction is not non-specific for DBDs. Lastly, we found that DNA-PK copurified with full-length human PR transiently expressed in HeLa cells, suggesting that the human PR/DNA-PK complex can assemble in vivo. These data show that DNA-PK and DBD(PR) interact, that DBD(PR) is a phosphorylation substrate of DNA-PK, and suggest a potential role for DNA-PK in PR-mediated transcription.


Assuntos
Antígenos Nucleares , DNA Helicases , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Progesterona/metabolismo , Proteínas de Saccharomyces cerevisiae , Sequência de Aminoácidos , Autoantígenos/isolamento & purificação , Autoantígenos/metabolismo , Sítios de Ligação , Proteína Quinase Ativada por DNA , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/isolamento & purificação , Glutationa Transferase/genética , Células HeLa , Humanos , Autoantígeno Ku , Ligantes , Metionina/metabolismo , Dados de Sequência Molecular , Fosforilação , Poli(ADP-Ribose) Polimerases/química , Poli(ADP-Ribose) Polimerases/isolamento & purificação , Biossíntese de Proteínas , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/isolamento & purificação , Receptores de Progesterona/química , Receptores de Progesterona/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Transcrição/metabolismo
10.
J Biol Chem ; 275(26): 20061-8, 2000 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-10751396

RESUMO

Steroid receptors represent a class of transcription regulators that act in part by overcoming the often repressive nature of chromatin to modulate gene activity. The mouse mammary tumor virus (MMTV) promoter is a useful model for studying transcriptional regulation by steroid hormone receptors in the context of chromatin. The chromatin architecture of the promoter prevents the assembly of basal transcription machinery and binding of ubiquitous transcription factors. However, in human breast carcinoma T47D cells lacking the glucocorticoid receptor (GR), but expressing the progesterone receptor (PR), nucleosome B (nuc B) assumes a constitutively hypersensitive chromatin structure. This correlation led us to test the hypothesis that the chromatin structure of nuc B was dependent on GR expression in T47D cells. To examine this possibility, we stably co-transfected the MMTV promoter and the GR into T47D cells that lacked both the GR and the PR. We found that in T47D cells that lack both the GR and the PR or express only the GR, nuc B assumes a constitutively "open" chromatin structure, which allows hormone independent access by restriction endonucleases and transcription factors. These results suggest that in GR(+)/pr(-) T47D cells, the MMTV chromatin structure permits GR transcriptional activation, independent of chromatin remodeling.


Assuntos
Neoplasias da Mama/virologia , Proteínas Estimuladoras de Ligação a CCAAT , Cromatina/metabolismo , Vírus do Tumor Mamário do Camundongo/genética , Regiões Promotoras Genéticas , Receptores de Glucocorticoides/metabolismo , Fatores de Transcrição , Western Blotting , Núcleo Celular/metabolismo , Cromatina/química , Proteínas de Ligação a DNA/metabolismo , Dexametasona/farmacologia , Glucocorticoides/farmacologia , Humanos , Hipersensibilidade , Fatores de Transcrição NFI , Proteínas Nucleares , Testes de Precipitina , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica , Transfecção , Células Tumorais Cultivadas , Proteína 1 de Ligação a Y-Box
11.
J Biol Chem ; 275(10): 7313-20, 2000 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-10702302

RESUMO

The role of the N-terminal region in nuclear receptor function was addressed by a biochemical and biophysical analysis of the progesterone receptor A-isoform lacking only the hormone binding domain (NT-A). Sedimentation studies demonstrate that NT-A is quantitatively monomeric, with a highly asymmetric shape. Contrary to dogma, the N-terminal region is structured as demonstrated by limited proteolysis. However, N-terminal structure is strongly stabilized by the DNA binding domain, possibly explaining the lack of structure seen in isolated activation domains. Upon DNA binding, NT-A undergoes N-terminal mediated assembly, suggestive of DNA-induced allostery, and consistent with changes in protease accessibility of sites outside the DNA binding domain. Microsequencing reveals that protease-accessible regions are limited to previously identified phosphorylation motifs and to functional domain boundaries.


Assuntos
DNA/metabolismo , Receptores de Progesterona/química , Sítios de Ligação , Endopeptidases/farmacologia , Humanos , Peso Molecular , Dobramento de Proteína , Elementos de Resposta , Transcrição Gênica
12.
Proc Natl Acad Sci U S A ; 97(3): 1032-7, 2000 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-10655479

RESUMO

Ligand-dependent down-regulation that leads to rapid and extensive loss of protein is characteristic of several nuclear steroid receptors, including human progesterone receptors (PRs). In breast cancer cells, >95% of PRs are degraded 6 h after the start of progestin treatment. The mechanism for down-regulation is unknown. We examined the role of PR phosphorylation by mitogen-activated protein kinases (MAPKs) in this process. Lactacystin and calpain inhibitor I, specific inhibitors of the 26S proteasome, blocked progestin-induced down-regulation, and ubiquitinated conjugates of PR accumulated in cells. Ligand-dependent PR degradation was also blocked by specific inhibition of p42 and p44 MAPKs. To define the targets of phosphorylation by this kinase, two serine/proline MAPK consensus sites on PR were mutated. We demonstrate that mutation of PR serine-294 to alanine (S294A) specifically and completely prevents ligand-dependent receptor down-regulation. We also find that rapid, ligand-independent degradation of immature PR intermediates occurs by a proteasome-mediated pathway. These results demonstrate that PR destruction, by either of two alternate routes, is mediated by the 26S proteasome. Specifically, down-regulation of mature PRs occurs by a mechanism in which ligand binding activates PR phosphorylation by MAPKs at a unique serine residue, which then targets the receptors for degradation.


Assuntos
Cisteína Endopeptidases/metabolismo , Regulação para Baixo/fisiologia , Sistema de Sinalização das MAP Quinases , Complexos Multienzimáticos/metabolismo , Proteínas de Neoplasias/metabolismo , Fosfosserina/química , Isoformas de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional , Receptores de Progesterona/metabolismo , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Neoplasias da Mama/química , Sequência Consenso , Cisteína Endopeptidases/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Feminino , Glicoproteínas/farmacologia , Células HeLa/metabolismo , Humanos , Imidazóis/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Complexos Multienzimáticos/efeitos dos fármacos , Fosforilação , Promegestona/farmacologia , Complexo de Endopeptidases do Proteassoma , Piridinas/farmacologia , Receptores de Progesterona/química , Proteínas Recombinantes de Fusão/metabolismo , Ubiquitinas/metabolismo
13.
J Steroid Biochem Mol Biol ; 74(5): 255-9, 2000 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-11162933

RESUMO

The antiestrogen tamoxifen is an effective treatment for estrogen receptor positive breast cancers, slowing tumor growth and preventing disease recurrence, with relatively few side effects. However, many patients who initially respond to treatment, later become resistant to treatment. Tamoxifen has both agonist and antagonist activities, which are manifested in a tissue-specific pattern. Development of tamoxifen resistance can be characterized by an increase in the partial agonist properties of the antiestrogen in the breast, resulting in loss of growth inhibition and even inappropriate tumor stimulation. Nuclear receptor function is modulated by transcriptional coregulators, which either enhance or repress receptor activity. Using a mixed antagonist-biased two-hybrid screening strategy, we identified two such proteins: the human homolog of the nuclear receptor corepressor, N-CoR, and a novel coactivator, L7/SPA (Switch Protein for Antagonists). In transcriptional studies N-CoR suppressed the agonist properties of tamoxifen and RU486, while L7/SPA increased agonist effects. We speculated that the relative level of these coactivators and corepressors might determine the balance of agonist and antagonist properties of mixed antagonists such as tamoxifen. Using quantitative RT-PCR we therefore measured the levels of transcripts encoding these coregulators, as well as the corepressor SMRT, and the coactivator SRC-1, in a small cohort of tamoxifen resistant and sensitive breast tumors. The results suggest that tumor sensitivity to mixed antagonists may be governed by a complex set of transcription factors, which we are only now beginning to understand.


Assuntos
Neoplasias da Mama/metabolismo , Resistencia a Medicamentos Antineoplásicos , Moduladores de Receptor Estrogênico/farmacologia , Tamoxifeno/farmacologia , Fatores de Transcrição/metabolismo , Animais , Neoplasias da Mama/tratamento farmacológico , Divisão Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Moduladores de Receptor Estrogênico/uso terapêutico , Feminino , Histona Acetiltransferases , Humanos , Mifepristona/farmacologia , Correpressor 2 de Receptor Nuclear , Coativador 1 de Receptor Nuclear , Receptores de Estrogênio/agonistas , Receptores de Estrogênio/antagonistas & inibidores , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/antagonistas & inibidores , Receptores de Progesterona/metabolismo , Proteínas Repressoras/metabolismo , Tamoxifeno/uso terapêutico
15.
J Steroid Biochem Mol Biol ; 69(1-6): 45-50, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10418980

RESUMO

Pharmacological antagonists of steroid receptor action had been thought to exert their effects by a passive mechanism driven principally by the ability of the antagonist to compete with agonist for the ligand binding site. However, recent analyses of antagonist-occupied receptor function suggest a more complex picture. Antagonists can be subdivided into two groups, type I, or pure antagonists, and type II, or mixed antagonists that can have variable transcriptional activity based upon differential dimerization and DNA binding properties. This led us to propose that receptor antagonism may not simply be a passive competition for the ligand binding site, but may, in some cases, involve active recruitment of corepressor or coactivator proteins to produce a mixed transcriptional phenotype. We used a yeast two-hybrid screen to identify proteins that interact specifically with antagonist-occupied receptors. Two proteins have been characterized: L7/SPA, a ribosome-associated protein that is localized in both the cytoplasm and nucleus, but with no known extranucleolar nuclear function; and hN-CoR, the human homolog of the mouse thyroid receptor corepressor mN-CoR. In in vivo transcription assays we show that L7/SPA enhances the partial agonist activity of type II mixed antagonists, and that N-CoR and the related corepressor, SMRT, suppresses it. The coregulators do not affect agonists or pure antagonists. Moreover, the net agonist activity seen with mixed antagonists is a function of the ratio of coactivator to corepressor. Based upon these results, we proposed that in breast tumors the inappropriate agonist activity seen with therapeutic antagonists such as tamoxifen is responsible for the hormone-resistant state. To confirm this, we are quantitating coactivator/corepressor ratios in breast tumor cells lines and clinical breast cancers. Results should provide new insights into the mechanisms underlying the progression of breast cancer to hormone resistance, and may suggest strategies for delaying or reversing this process.


Assuntos
Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/fisiopatologia , Receptores de Esteroides/antagonistas & inibidores , Tamoxifeno/farmacologia , Transcrição Gênica , Animais , Resistencia a Medicamentos Antineoplásicos , Humanos , Receptores de Esteroides/genética
16.
Mol Endocrinol ; 13(6): 829-36, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10379882

RESUMO

In the breast, data from numerous laboratories suggest that cross-talk exists between PR and growth factor and cytokine signaling pathways at multiple levels (Fig. 4). At the cell surface (level 1), progestins up-regulate growth factor and cytokine receptors. We have expanded this observation by examining the effects of progestins in the cytoplasm (level 2) where progestins regulate several intracellular effectors by increasing the levels and altering the subcellular compartmentalization of Stat5, increasing the association of Stat5 with phosphotyrosine-containing proteins and tyrosine phosphorylation of JAK2, Cbl, and Shc, and potentiating EGF-stimulated p42/p44 MAPKs, p38 MAP kinase, and JNK activities. Together, these events lead to sensitization of downstream signaling pathways to the actions of locally acting secondary factors. Finally, inside the nucleus (level 3), agonist-occupied PR synergize with nuclear transcription factors that are growth-factor regulated, to control the activity of key genes involved in breast cell fate (Figs. 1 and 4). We speculate that after progesterone treatment, orchestrated combinations of steroid hormones and growth factors or cytokines can fine tune the timing and degree of expression of a subset of genes that determine whether progestin-primed cells undergo proliferation, differentiation, or programmed cell death. The paradoxical effects of progesterone have presented a longstanding conundrum to the scientist and clinician. Why are physiological levels of progesterone proliferative in the breast but antiproliferative and protective in the uterus? If progesterone is proliferative in the breast, why is high-dose progestin therapy successful in treating breast cancer? Our intent here has been to open a dialogue addressing these questions. Our data and that of others are beginning to show that one cannot approach the question of progestin actions in isolation. Other important regulatory proteins, whose expression may vary in tissue-specific ways, work in concert with progesterone to decide cell fate. The timing and dose of progesterone may also influence the biological response. Since progestins are widely used in oral contraception, in hormone replacement therapy, and in cancer treatments, it is becoming critically important that the subtleties of their mechanisms of action be clearly understood.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proteínas do Leite , Progesterona/metabolismo , Transdução de Sinais , Animais , Neoplasias da Mama/tratamento farmacológico , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Ciclo Celular/efeitos dos fármacos , Divisão Celular , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , Ciclinas/metabolismo , Citocinas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta a Droga , Feminino , Regulação Neoplásica da Expressão Gênica , Substâncias de Crescimento/metabolismo , Humanos , Progesterona/farmacologia , Fator de Transcrição STAT5 , Transativadores/metabolismo , Fatores de Transcrição/metabolismo
17.
Circulation ; 99(20): 2688-93, 1999 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-10338464

RESUMO

BACKGROUND: Estrogens stimulate growth of breast or uterine cells but have the opposite effect on vascular smooth muscle cells, in which they protect against coronary artery disease with or without concomitant administration of progesterone. A possible cause of differences in hormone action is variable tissue-specific expression of hormone receptor. Therefore, we analyzed the structure of estrogen receptors (ERs) and progesterone receptors (PRs) in human vascular smooth muscle. METHODS AND RESULTS: RNA was isolated from human vascular smooth muscle, and the functional domains of ER-alpha and PR were characterized by reverse transcriptase and polymerase chain reaction. Interestingly, in addition to wild-type ER-alpha and PR, 5 variant ER-alpha and 2 variant PR transcripts were found. These variants contained precise deletions of exons encoding regions of the hormone-binding domain. The PR transcripts lacked exon 4 (PRDelta4) and exon 6 (PRDelta6). The ER-alpha transcripts were missing exon 4 (ERDelta4), exon 5 (ERDelta5), exon 6 (ERDelta6), exon 7 (ERDelta7), and exons 6 and 7, (ERDelta6,7). ER-beta variants were also detected. The PR variants were functionally characterized, and PRDelta6 was found to be a dominant-negative transcription inhibitor of wild-type receptors. Variant PR was present in premenopausal women but absent in postmenopausal women. CONCLUSIONS: Variant PR and ER transcripts are extensively expressed in human vascular smooth muscle. The complex tissue-specific effects of sex hormones may be mediated by the expression of heterogeneous forms of their cognate receptors. The presence of variant ERs and PRs may be of importance in altering the physiological effects of estrogens or progestins in vascular smooth muscle.


Assuntos
Variação Genética , Músculo Liso Vascular/metabolismo , RNA Mensageiro/metabolismo , Receptores de Estrogênio/genética , Receptores de Progesterona/genética , Adulto , Idoso , Técnicas de Cultura , Receptor beta de Estrogênio , Éxons/genética , Feminino , Deleção de Genes , Variação Genética/fisiologia , Humanos , Masculino , Pessoa de Meia-Idade , Pós-Menopausa/metabolismo , Pré-Menopausa/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
J Biol Chem ; 273(47): 31308-16, 1998 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-9813039

RESUMO

During late stages of breast cancer progression, tumors frequently acquire steroid hormone resistance with concurrent amplification of growth factor receptors; this alteration predicts a poor prognosis. We show here that following treatment with the progestin, R5020, breast cancer cells undergo a "biochemical shift" in the regulation of epidermal growth factor (EGF)-stimulated signaling pathways: R5020 potentiates the effects of EGF by up-regulating EGFR, c-ErbB2 and c-ErbB3 receptors, and by enhancing EGF-stimulated tyrosine phosphorylation of signaling molecules known to associate with activated type I receptors. Independently of EGF, R5020 increases Stat5 protein levels, association of Stat5 with phosphotyrosine-containing proteins, and tyrosine phosphorylation of JAK2 and Shc. Furthermore, progestins "prime" breast cancer cells for growth signals by potentiating EGF-stimulated p42/p44 mitogen-activated protein kinase (MAPK), p38 MAP kinase, and JNK activities. Although the levels of cyclin D1, cyclin E, and p21(WAF1), are up-regulated by R5020 alone, they are synergistically up-regulated by EGF in the presence of R5020. Up-regulation of cell cycle proteins by EGF is blocked by inhibition of p42/p44 MAPK only in the presence of R5020, supporting a shift in the regulation of these cell cycle mediators from MAPK-independent to MAPK-dependent pathways. In summary, progesterone selectively increases the sensitivity of key kinase cascades to growth factors, thereby priming cells for stimulation by latent growth signals. These data support a model in which breast cancer cell growth switches from steroid hormone to growth factor dependence.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Proteínas Quinases Ativadas por Mitógeno , Progesterona/metabolismo , Receptor Cross-Talk , Ciclo Celular/fisiologia , Linhagem Celular , Feminino , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Modelos Biológicos , Fosfotirosina/metabolismo , Promegestona/farmacologia , Receptores de Fatores de Crescimento/biossíntese , Transdução de Sinais , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA