Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Breast Cancer Res ; 19(1): 79, 2017 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-28673316

RESUMO

BACKGROUND: Upregulation of estrogen receptor beta (ERß) in breast cancer cells is associated with epithelial maintenance, decreased proliferation and invasion, and a reduction in the expression of the receptor has been observed in invasive breast tumors. However, proof of an association between loss of ERß and breast carcinogenesis is still missing. METHODS: To study the role of ERß in breast oncogenesis, we generated mouse conditional mutants with specific inactivation of ERß and p53 in the mammary gland epithelium. For epithelium-specific knockout of ERß and p53, ERß F/F and p53 F/F mice were crossed to transgenic mice that express the Cre recombinase under the control of the human keratin 14 promoter. RESULTS: Somatic loss of ERß significantly accelerated formation of p53-deficient mammary tumors. Loss of the receptor also resulted in the development of less differentiated carcinomas with stronger spindle cell morphology and decreased expression of luminal epithelial markers. CONCLUSIONS: Our results show that synergism between ERß and p53 inactivation functions to determine important aspects of breast oncogenesis and cancer progression.


Assuntos
Neoplasias da Mama/genética , Transformação Celular Neoplásica/genética , Receptor beta de Estrogênio/genética , Proteína Supressora de Tumor p53/genética , Alelos , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/metabolismo , Transição Epitelial-Mesenquimal/genética , Epitélio/metabolismo , Receptor beta de Estrogênio/metabolismo , Feminino , Expressão Gênica , Inativação Gênica , Genótipo , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Camundongos , Camundongos Transgênicos , Fenótipo , Prognóstico , Proteína Supressora de Tumor p53/metabolismo
2.
Am J Physiol Renal Physiol ; 309(2): F98-108, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-26017975

RESUMO

TNF has been implicated in glomerular diseases, but its actions on podocytes are not well understood. Endogenous TNF expression is markedly increased in mouse podocytes exposed to sera from patients with recurrent focal segmental glomerulosclerosis, and TNF is able to increase its own expression in these cells. Exposure of podocytes to TNF increased phosphorylation of NF-κB p65-RelA followed by increased tyrosine phosphorylation of STAT3. STAT3 activation was blocked by the NF-κB inhibitor JSH-23 and by the STAT3 inhibitor stattic, whereas TNF-evoked NF-κB activation was not affected by stattic. TNF treatment increased nuclear accumulation of nuclear factor of activated T cells (NFAT)c1 in podocytes, a process that occurred downstream of STAT3 activation. TNF also increased expression of cyclin D1 but had no effect on cyclin-dependent kinase 4, p27(kip), or podocin. Despite its effects on cyclin D1, TNF treatment for up to 72 h did not cause podocytes to reenter the cell cycle. TNF increased total expression of transient receptor potential (TRP)C6 channels through a pathway dependent on NFATc1 and increased the steady-state expression of TRPC6 subunits on the podocyte cell surface. TNF effects on TRPC6 trafficking required ROS. Consistent with this, La(3+)-sensitive cationic currents activated by a diacylglycerol analog were increased in TNF-treated cells. The effects of TNF on NFATc1 and TRPC6 expression were blocked by cyclosporine A but were not blocked by the pan-TRP inhibitor SKF-96365. TNF therefore influences multiple pathways previously implicated in podocyte pathophysiology and is likely to sensitize these cells to other insults.


Assuntos
Podócitos/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo , Adulto , Animais , Linhagem Celular , Glomerulosclerose Segmentar e Focal/metabolismo , Humanos , Masculino , Camundongos , Fatores de Transcrição NFATC/metabolismo , Fator de Transcrição STAT3/metabolismo , Canais de Cátion TRPC/metabolismo , Canal de Cátion TRPC6
3.
Breast Cancer Res ; 14(6): R148, 2012 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-23158001

RESUMO

INTRODUCTION: Epithelial to mesenchymal transition (EMT) is associated with the basal-like breast cancer phenotypes. 60% of basal-like cancers have been shown to express wild-type estrogen receptor beta (ERbeta1). However, it is still unclear whether the ERbeta expression is related to EMT, invasion and metastasis in breast cancer. In the present study, we examined whether ERbeta1 through regulating EMT can influence invasion and metastasis in basal-like cancers. METHODS: Basal-like breast cancer cells (MDA-MB-231 and Hs578T) in which ERbeta1 was either overexpressed or downregulated were analyzed for their ability to migrate and invade (wound-healing assay, matrigel-coated Transwell assay) as well as for the expression of EMT markers and components of the EGFR pathway (immunoblotting, RT-PCR). Coimmunoprecipitation and ubiquitylation assays were employed to examine whether ERbeta1 alters EGFR protein degradation and the interaction between EGFR and the ubiquitin ligase c-Cbl. The metastatic potential of the ERbeta1-expressing MDA-MB-231 cells was evaluated in vivo in a zebrafish xenotransplantation model and the correlation between ERbeta1 and E-cadherin expression was examined in 208 clinical breast cancer specimens by immunohistochemistry. RESULTS: Here we show that ERbeta1 inhibits EMT and invasion in basal-like breast cancer cells when they grow either in vitro or in vivo in zebrafish. The inhibition of EMT correlates with an ERbeta1-mediated upregulation of miR-200a/b/429 and the subsequent repression of ZEB1 and SIP1, which results in increased expression of E-cadherin. The positive correlation of ERbeta1 and E-cadherin expression was additionally observed in breast tumor samples. Downregulation of the basal marker EGFR through stabilization of the ubiquitin ligase c-Cbl complexes and subsequent ubiquitylation and degradation of the activated receptor is involved in the ERbeta1-mediated repression of EMT and induction of EGFR signaling abolished the ability of ERbeta1 to sustain the epithelial phenotype. CONCLUSIONS: Taken together, the results of our study strengthen the association of ERbeta1 with the regulation of EMT and propose the receptor as a potential crucial marker in predicting metastasis in breast cancer.


Assuntos
Neoplasias da Mama/patologia , Transição Epitelial-Mesenquimal , Receptores ErbB/metabolismo , Receptor beta de Estrogênio/metabolismo , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Caderinas/biossíntese , Linhagem Celular Tumoral , Receptor beta de Estrogênio/biossíntese , Receptor beta de Estrogênio/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/biossíntese , Humanos , Células MCF-7 , MicroRNAs/biossíntese , Invasividade Neoplásica/patologia , Metástase Neoplásica/patologia , Transplante de Neoplasias , Neoplasia de Células Basais , Proteínas do Tecido Nervoso/biossíntese , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Proteínas de Ligação a RNA/biossíntese , Proteínas Repressoras/biossíntese , Fatores de Transcrição da Família Snail , Fatores de Transcrição/biossíntese , Fatores de Transcrição/metabolismo , Transplante Heterólogo , Ubiquitinação , Peixe-Zebra , Homeobox 2 de Ligação a E-box com Dedos de Zinco , Homeobox 1 de Ligação a E-box em Dedo de Zinco
5.
Breast Cancer Res Treat ; 127(2): 417-27, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20623183

RESUMO

Estrogen receptor beta (ERß) inhibits proliferation in different cellular systems by regulating components of the cell cycle machinery. Eukaryotic cells respond to DNA damage by arresting in G1, S, or G2 phases of the cell cycle to initiate DNA repair. Most tumor cells due to disruptions in the p53-dependent G1 pathway are dependent on S-phase and G2/M checkpoints to maintain genomic integrity in response to DNA damage. We report that induction of ERß expression causes abrogation of the S-phase, and the Chk1/Cdc25C-mediated G2/M checkpoints after cisplatin and doxorubicin exposure in p53-defective breast cancer cells but not in p53 wild-type mammary cells. This impairment of DNA damage response that involves BRCA1 downregulation and caspase-2 activation results in mitotic catastrophe and decreased cancer cell survival. These results indicate that in cancers where p53 is defective, assessment of the presence of ERß may be of predictive value for the successful response to chemotherapy.


Assuntos
Ciclo Celular/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Receptor beta de Estrogênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Proteína Supressora de Tumor p53/deficiência , Antineoplásicos/farmacologia , Proteína BRCA1/metabolismo , Ciclo Celular/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Cisplatino/farmacologia , Dano ao DNA/genética , Células HeLa , Humanos , Ligantes , Mitose/efeitos dos fármacos , Mitose/genética , Proteína Supressora de Tumor p53/genética
6.
Cancer Res ; 81(9): 2399-2414, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33514514

RESUMO

Inflammatory breast cancer (IBC) is a highly metastatic breast carcinoma with high frequency of estrogen receptor α (ERα) negativity. Here we explored the role of the second ER subtype, ERß, and report expression in IBC tumors and its correlation with reduced metastasis. Ablation of ERß in IBC cells promoted cell migration and activated gene networks that control actin reorganization, including G-protein-coupled receptors and downstream effectors that activate Rho GTPases. Analysis of preclinical mouse models of IBC revealed decreased metastasis of IBC tumors when ERß was expressed or activated by chemical agonists. Our findings support a tumor-suppressive role of ERß by demonstrating the ability of the receptor to inhibit dissemination of IBC cells and prevent metastasis. On the basis of these findings, we propose ERß as a potentially novel biomarker and therapeutic target that can inhibit IBC metastasis and reduce its associated mortality. SIGNIFICANCE: These findings demonstrate the capacity of ERß to elicit antimetastatic effects in highly aggressive inflammatory breast cancer and propose ERß and the identified associated genes as potential therapeutic targets in this disease.


Assuntos
Actinas/metabolismo , Movimento Celular/genética , Receptor beta de Estrogênio/metabolismo , Neoplasias Inflamatórias Mamárias/metabolismo , Transdução de Sinais/genética , Citoesqueleto de Actina/metabolismo , Animais , Estudos de Coortes , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/genética , Feminino , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Neoplasias Inflamatórias Mamárias/genética , Neoplasias Inflamatórias Mamárias/patologia , Células MCF-7 , Camundongos , Metástase Neoplásica/genética , Transfecção , Carga Tumoral/genética , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Oncogene ; 39(22): 4344-4357, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32335582

RESUMO

We explore a novel strategy of activating immune signaling through increased micronuclei formation utilizing a cell cycle checkpoint inhibitor to drive cell cycle progression following ionizing radiation. The Chk1/2 inhibitor AZD7762 is used to abrogate radiation therapy (RT)-induced G2/M cell cycle arrest in multiple cell lines and, we find that this therapeutic combination promotes increased micronuclei formation in vitro and subsequently drives increased type I interferon signaling and cytotoxic T-cell activation. In vivo studies using B16-F10 melanoma cancer cells implanted in C57/BL6 mice demonstrate improved rates of tumor control at the abscopal (unirradiated) site, located outside of the radiation field, only in the AZD7762 + RT group, with a corresponding reduction in mean tumor volume, increase in the CD8 T-cell population, and immune activated gene signaling. Our results demonstrate that targeted inhibition of cell cycle checkpoint activation following ionizing radiation drives increased production of immunogenic micronuclei, leading to systemic tumor response with potential future clinical benefit.


Assuntos
Núcleo Celular/efeitos dos fármacos , Núcleo Celular/efeitos da radiação , Quinase 1 do Ponto de Checagem/antagonistas & inibidores , Quinase do Ponto de Checagem 2/antagonistas & inibidores , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos da radiação , Melanoma Experimental/imunologia , Proteínas de Neoplasias/antagonistas & inibidores , Tiofenos/farmacologia , Ureia/análogos & derivados , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Interferon beta/biossíntese , Interferon beta/genética , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/patologia , Melanoma Experimental/radioterapia , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Testes para Micronúcleos , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Fator de Transcrição STAT1/biossíntese , Fator de Transcrição STAT1/genética , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/efeitos da radiação , Ureia/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Oncogene ; 38(12): 2092-2107, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30442981

RESUMO

Aberrant Notch signaling is implicated in several cancers, including breast cancer. However, the mechanistic details of the specific receptors and function of ligand-mediated Notch signaling that promote breast cancer remains elusive. In our studies we show that DLL1, a Notch signaling ligand, is significantly overexpressed in ERα+ luminal breast cancer. Intriguingly, DLL1 overexpression correlates with poor prognosis in ERα+ luminal breast cancer, but not in other subtypes of breast cancer. In addition, this effect is specific to DLL1, as other Notch ligands (DLL3, JAGGED1, and JAGGED2) do not influence the clinical outcome of ERα+ patients. Genetic studies show that DLL1-mediated Notch signaling in breast cancer is important for tumor cell proliferation, angiogenesis, and cancer stem cell function. Consistent with prognostic clinical data, we found the tumor-promoting function of DLL1 is exclusive to ERα+ luminal breast cancer, as loss of DLL1 inhibits both tumor growth and lung metastasis of luminal breast cancer. Importantly, we find that estrogen signaling stabilizes DLL1 protein by preventing its proteasomal and lysososmal degradations. Moreover, estrogen inhibits ubiquitination of DLL1. Together, our results highlight an unexpected and novel subtype-specific function of DLL1 in promoting luminal breast cancer that is regulated by estrogen signaling. Our studies also emphasize the critical role of assessing subtype-specific mechanisms driving tumor growth and metastasis to generate effective subtype-specific therapeutics.


Assuntos
Neoplasias da Mama/patologia , Estrogênios/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Animais , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/metabolismo , Proteínas de Ligação ao Cálcio , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica , Progressão da Doença , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Lisossomos/metabolismo , Camundongos , Metástase Neoplásica , Células-Tronco Neoplásicas/patologia , Neovascularização Patológica , Prognóstico , Ubiquitinação
9.
Curr Ther Res Clin Exp ; 69(5): 423-39, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24692817

RESUMO

BACKGROUND: Several antioxidants have been found to have conflicting results in attenuating myocardial reperfusion injury. These studies were done primarily in experimental protocols that did not approximate clinical situations. OBJECTIVE: The aim of this study was to test the efficacy of 3 different antioxidants (ascorbic acid [AA], desferrioxamine, and N-acetylcysteine [NAC]) administered IV alone and in combination in a closed-chest pig model. METHODS: Farm-raised domestic male pigs (aged 3-5 months, weight of 30-35 kg) were assigned to 1 of 5 groups to receive treatment as follows: group A, AA 100 mg/kg; group B, desferrioxamine 60 mg/kg; group C, a loading dose of NAC 100 mg/kg for 20 minutes and a 20-mg/kg maintenance dose; group D, all 3 drugs in combination; and group E, normal saline (control group). The infusion of all drugs was started 15 minutes before and completed 5 minutes after reperfusion, except for the administration of NAC, which was terminated 60 minutes postreperfusion. Myocardial ischemia (45 minutes) and reperfusion (210 minutes) were achieved percutaneously by circumflex artery balloon occlusion. Ejection fraction, left ventricular end-diastolic pressure (LVEDP), flow in the infarcted artery, and all ventricular arrhythmias were recorded. Oxidative stress was estimated by serial measurements of thiobarbituric acid reactive substance (TBARS) concentration in coronary sinus blood. Infarct size was assessed as a percentage of the area at risk (I/R ratio) using the tetrazolium red staining method. RESULTS: The 25 pigs were divided into 5 groups of 5 pigs each. No significant between-group differences were found in I/R ratio or in oxidative stress (as measured by TBARS concentration). Group C developed significantly more ventricular atrhythmias than the control group (80% vs 0%, P = 0.02). No other differences among groups were found. LVEDP was significantly elevated in all treatment groups (mean LVEDP difference [SD] for group A, 6.0 [1.6] mm Hg; group B, 17.6 [1.9] mm Hg; group C, 3.6 [1.7] mm Hg; group D, 6.8 [3.2] and group E, 5.4 [3.4] mm Hg). LVEDP elevation was found to be significantly higher in group B compared with all the other groups (all, P < 0.001). No significant between-group differences were found in the other parameters measured. CONCLUSION: In this experimental pig model, the antioxidants AA, desferrioxamine, and NAC administered alone or in combination did not reduce the deleterious effects of reperfusion injury and specifically the extent of myocardial necrosis.

11.
Mol Cancer Res ; 16(2): 233-242, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29117942

RESUMO

The expression of wild-type estrogen receptor ß (ESR2/ERß1) correlates with clinical outcome in patients with non-small cell lung cancer (NSCLC). However, the molecular mechanism that accounts for this association is currently poorly understood. ERß1 was previously linked to chemotherapy response in patients with breast cancer and in breast cancer cells. The effect of the receptor in NSCLC cells after chemotherapy treatment, a common remedy for advanced NSCLC, has not been studied. Here, upregulation of ERß1 increases the sensitivity of NSCLC cells to treatment with doxorubicin and etoposide. This effect was primarily observed in p53-defecient NSCLC cells. In these cells, ERß1 either enhanced G2-M cell-cycle arrest by activating the checkpoint kinase 1 (Chk1) and altering downstream signaling or induced apoptosis. The expression of p63 target genes that control G2-M checkpoint activation was altered by ERß1 suggesting an ERß1-p63 transcriptional cooperation in lung cancer cells that affects DNA damage response (DDR). These results suggest involvement of ERß1 in the mechanism that regulates DNA damage response in NSCLC cells and support the potential predictive and therapeutic value of the receptor in clinical management of the disease.Implications: This study demonstrating the impact of ERß1 on chemosensitivity of NSCLC cells suggests the predictive value of the receptor for successful response of tumors to chemotherapy and the potential benefit of chemotherapy-treated patients from the use of ER ligands. Mol Cancer Res; 16(2); 233-42. ©2017 AACR.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Doxorrubicina/farmacologia , Receptor beta de Estrogênio/genética , Etoposídeo/farmacologia , Neoplasias Pulmonares/genética , Regulação para Cima , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Dano ao DNA , Reparo do DNA/efeitos dos fármacos , Receptor beta de Estrogênio/metabolismo , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Transfecção , Proteína Supressora de Tumor p53/genética
12.
Oncotarget ; 9(32): 22509-22522, 2018 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-29854295

RESUMO

Estrogen receptor α (ERα)-positive breast cancers tend to develop resistance to both endocrine therapy and chemotherapy. Despite recent progress in defining molecular pathways that confer endocrine resistance, the mechanisms that regulate chemotherapy response in luminal tumors remain largely elusive. Luminal tumors often express wild-type p53 that is a major determinant of the cellular DNA damage response. Similar to p53, the second ER subtype, ERß, has been reported to inhibit breast tumorigenesis by acting alone or in collaboration with p53. However, a synergistic mechanism of action has not been described. Here, we suggest that ERß relies on p53 to elicit its tumor repressive actions in ERα-positive breast cancer cells. Upregulation of ERß and treatment with ERß agonists potentiates the tumor suppressor function of p53 resulting in decreased survival. This effect requires molecular interaction between the two proteins that disrupts the inhibitory action of ERα on p53 leading to increased transcriptional activity of p53. In addition, we show that the same interaction alters the chemosensitivity of endocrine-resistant cells including their response to tamoxifen therapy. Our results suggest a collaboration of ERß and p53 tumor suppressor activity in breast cancer cells that indicates the importance of ligand-regulated ERß as a tool to target p53 activity and improve the clinical management of resistant disease.

13.
J Steroid Biochem Mol Biol ; 163: 45-50, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27045680

RESUMO

Activation of the unfolded protein response (UPR) confers resistance to anti-estrogens and chemotherapeutics in estrogen receptor α (ERα)-positive and triple-negative breast cancers. Among the regulators of the UPR in breast cancer is estrogen signaling. Estrogen regulates major components of the UPR and ER expression is associated with the sensitivity of tumor cells to UPR-regulated apoptosis. Recent studies have confirmed the crosstalk between the ERs and UPR and suggest novel therapeutic strategies that combine targeting of both signaling pathways. These remedies may be more effective in repressing oncogenic adaptive mechanisms and benefit patients with resistant disease.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias da Mama/metabolismo , Antagonistas de Estrogênios/uso terapêutico , Estrogênios/metabolismo , Regulação Neoplásica da Expressão Gênica , Transdução de Sinais/genética , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Fator 6 Ativador da Transcrição/genética , Fator 6 Ativador da Transcrição/metabolismo , Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Feminino , Humanos , Proteína 1 de Ligação a X-Box/genética , Proteína 1 de Ligação a X-Box/metabolismo , eIF-2 Quinase/genética , eIF-2 Quinase/metabolismo
14.
Oncotarget ; 7(12): 13599-611, 2016 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-26871946

RESUMO

Most (80%) of the triple-negative breast cancers (TNBCs) express mutant p53 proteins that acquire oncogenic activities including promoting metastasis. We previously showed that wild-type ERß (ERß1) impedes epithelial to mesenchymal transition (EMT) and decreases the invasiveness of TNBC cells. In the present study we searched for signaling pathways that ERß1 uses to inhibit EMT and invasion in TNBC cells. We show that ERß1 binds to and opposes the transcriptional activity of mutant p53 at the promoters of genes that regulate metastasis. p63 that transcriptionally cooperates with mutant p53 also binds to ERß1. Downregulation of p63 represses the epithelial phenotype of ERß1-expressing cells and alters the expression of mutant p53 target genes. These results describe a novel mechanism through which ERß1 can disturb oncogenic signals to inhibit aggressiveness in TNBCs.


Assuntos
Transição Epitelial-Mesenquimal , Receptor beta de Estrogênio/metabolismo , Regulação Neoplásica da Expressão Gênica , Mutação , Neoplasias de Mama Triplo Negativas/patologia , Proteína Supressora de Tumor p53/genética , Apoptose , Movimento Celular , Proliferação de Células , Receptor beta de Estrogênio/genética , Humanos , Invasividade Neoplásica , Transdução de Sinais , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/metabolismo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo
15.
Trends Endocrinol Metab ; 26(9): 453-4, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26277479

RESUMO

Copy number loss of the PGR gene and decreased expression of progesterone receptor (PR) may account for worse clinical outcomes in some individuals with estrogen receptor α (ERα)-positive breast cancer. A recent report shows that PR activation inhibits estrogen-driven breast tumor growth by altering ERα chromatin binding and transcriptional activity.


Assuntos
Neoplasias da Mama/genética , Receptor Cross-Talk , Receptores de Estrogênio/genética , Receptores de Progesterona/genética , Receptor alfa de Estrogênio/genética , Feminino , Humanos
16.
Trends Endocrinol Metab ; 26(9): 467-76, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26183887

RESUMO

A significant number of estrogen receptor α (ERα)-positive breast tumors develop resistance to endocrine therapy and recur with metastatic disease. Several mechanisms of endocrine resistance have been proposed, including genetic alterations that lead to ERs with altered protein sequence. By altering the conformation of the protein and increasing the interaction with coactivators, point mutations in ESR1, the gene encoding ERα, promote an active form of the receptor in the absence of hormone that assists tumor cells to evade hormonal treatments. Recent studies have confirmed that ESR1 point mutations frequently occur in metastatic breast tumors that are refractory to endocrine therapy, and suggest the development of novel strategies that may be more effective in controlling ER signaling and benefit patients with recurrent and metastatic disease.


Assuntos
Neoplasias da Mama/genética , Receptor alfa de Estrogênio/genética , Receptores de Estrogênio/genética , Neoplasias da Mama/epidemiologia , Feminino , Humanos , Mutação/genética
17.
Mol Cancer Res ; 12(6): 843-54, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24618619

RESUMO

UNLABELLED: Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related deaths worldwide. In addition to the aberrant growth factor signaling, dysregulation of other pathways, such as those mediated by estrogens and their receptors, has been linked to NSCLC initiation and progression. Although the expression of wild-type estrogen receptor ß (ERß1) has been associated with prolonged disease-free survival in patients with NSCLC, the molecular mechanism that accounts for this correlation is unknown. Here, upregulation of ERß1 reduced proliferation and enhanced apoptosis in the context of mutant RAS. ERß1 was found to induce apoptosis by stimulating the intrinsic apoptotic pathway that involves BIM, a Bcl-2 proapoptotic family member that is regulated by the extracellular signal-regulated kinase (ERK). Downregulation of EGFR and inactivation of RAS and the downstream components ERK1/2 were found to be involved in the ERß1-induced apoptosis. Manipulation of EGFR and RAS expression and activity in ERß1-expressing cells revealed the central role of oncogenic RAS inhibition in the ERß1-mediated proapoptotic phenotype and EGFR regulation. These results demonstrate that ERß1 decreases the survival of NSCLC cells by regulating oncogenic RAS signaling. IMPLICATIONS: The ability of ERß1 to regulate the oncogenic functions of RAS suggests its importance in the biology of NSCLC and its clinical management. Mol Cancer Res; 12(6); 843-54. ©2014 AACR.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Receptor beta de Estrogênio/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas ras/metabolismo , Apoptose/fisiologia , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Progressão da Doença , Intervalo Livre de Doença , Receptor beta de Estrogênio/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Fenótipo , Transdução de Sinais , Transfecção , Proteínas ras/genética
18.
Oncotarget ; 5(11): 3871-9, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25003695

RESUMO

P53 mutations are associated with invasive tumors in mouse models. We assessed the p53mutations and survival in patients with advanced cancer treated in the Phase I Program. Of 691 tested patients, 273 (39.5%) had p53 mutations. Patients with p53 mutations were older (p<.0001) and had higher numbers of liver metastases (p=.005). P53 mutations were associated with higher numbers of other aberrations; PTEN (p=.0005) and HER2 (p=.003)aberrations were more common in the p53 mutation group. No survival difference was observed between patients with p53 mutations and those with wild-type p53. In patients with wild-type p53 and other aberrations, patients treated with matched-therapy against the additional aberrations had longer survival compared to those treated with non-matched-therapy or those who received no therapy (median survival, 26.0 vs. 11.8 vs. 9.8 months, respectively; p= .0007). Results were confirmed in a multivariate analysis (p= .0002). In the p53 mutation group with additional aberrations, those who received matched-therapy against the additional aberrations had survival similar to those treated with non-matched-therapy or those who received no therapy (p=.15). In conclusion, our results demonstrated resistance to matched-targeted therapy to the other aberrations in patients with p53 mutations and emphasize the need to overcome this resistance.


Assuntos
Genes p53 , Mutação , Neoplasias/genética , Ensaios Clínicos Fase I como Assunto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias/terapia , Prognóstico , Análise de Sobrevida , Taxa de Sobrevida , Resultado do Tratamento
19.
J Clin Invest ; 122(8): 2771-3, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22820293

RESUMO

Alteration of the ERα/ERß balance is a critical step in breast cancer development and progression, and selective restoration of the activity of estrogen receptors has been proposed as one of the major therapeutic approaches for breast cancer. In this issue of JCI, Cheng et al. show that, by differentially modulating the stability of ERα and ERß, PES1 increases the ERα/ERß ratio and triggers breast tumor growth. These findings highlight PES1 as a potential target for the treatment of breast cancer.

20.
Nat Rev Cancer ; 11(8): 597-608, 2011 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-21779010

RESUMO

By eliciting distinct transcriptional responses, the oestrogen receptors (ERs) ERα and ERß exert opposite effects on cellular processes that include proliferation, apoptosis and migration and that differentially influence the development and the progression of cancer. Perturbation of ER subtype-specific expression has been detected in various types of cancer, and the differences in the expression of ERs are correlated with the clinical outcome. The changes in the bioavailability of ERs in tumours, together with their specific biological functions, promote the selective restoration of their activity as one of the major therapeutic approaches for hormone-dependent cancers.


Assuntos
Receptor alfa de Estrogênio/fisiologia , Receptor beta de Estrogênio/fisiologia , Neoplasias/fisiopatologia , Animais , Humanos , Neoplasias/tratamento farmacológico , Prognóstico , Isoformas de Proteínas
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa