Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Proc Natl Acad Sci U S A ; 111(12): 4472-7, 2014 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-24567396

RESUMO

Mutations in the tumor suppressor BRCA1 predispose women to breast and ovarian cancers. The mechanism underlying the tissue-specific nature of BRCA1's tumor suppression is obscure. We previously showed that the antioxidant pathway regulated by the transcription factor NRF2 is defective in BRCA1-deficient cells. Reactivation of NRF2 through silencing of its negative regulator KEAP1 permitted the survival of BRCA1-null cells. Here we show that estrogen (E2) increases the expression of NRF2-dependent antioxidant genes in various E2-responsive cell types. Like NRF2 accumulation triggered by oxidative stress, E2-induced NRF2 accumulation depends on phosphatidylinositol 3-kinase-AKT activation. Pretreatment of mammary epithelial cells (MECs) with the phosphatidylinositol 3-kinase inhibitor BKM120 abolishes the capacity of E2 to increase NRF2 protein and transcriptional activity. In vivo the survival defect of BRCA1-deficient MECs is rescued by the rise in E2 levels associated with pregnancy. Furthermore, exogenous E2 administration stimulates the growth of BRCA1-deficient mammary tumors in the fat pads of male mice. Our work elucidates the basis of the tissue specificity of BRCA1-related tumor predisposition, and explains why oophorectomy significantly reduces breast cancer risk and recurrence in women carrying BRCA1 mutations.


Assuntos
Proteína BRCA1/genética , Sobrevivência Celular/fisiologia , Estrogênios/fisiologia , Fator 2 Relacionado a NF-E2/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Animais , Feminino , Xenoenxertos , Camundongos , Camundongos Transgênicos , Estresse Oxidativo
2.
Mod Pathol ; 27(7): 991-1001, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24336157

RESUMO

Alterations in the retinoblastoma pathway are frequent in ovarian/tubal high-grade serous cancers, but the mechanism of deregulation and the impact on patient outcome are poorly understood. A cohort of 334 high-grade serous carcinomas was studied by immunohistochemical analysis of RB1, p16, cyclin D1, cyclin E1, and Ki67. Additional detailed analyses including RB1 allelic deletion (n=42), mutation (n=75), methylation (n=31), and SNP array analyses (n=75) were performed on cases with clinical parameters, including age, debulking status, treatment, and clinical outcome. p16/RB1 expression results yielded three distinct clinically relevant subgroups upon multivariable analysis controlling for stage, debulking status, and treatment types: p16 homogeneous/RB1+ with the shortest progression-free survival (median 15 months (95% CI: 13-18); P=0.016) compared with the p16 heterogeneous/RB1+ subgroup (median 22 months (95% CI: 16-32)) and the p16 homogeneous/RB1- subgroup (median 20 months (95% CI: 15-24)). Patients in the p16 homo/RB1- subgroup showed a significant increase in overall survival (>60 months; P=0.013), which suggests an increase in sensitivity to cytotoxic agents. Analyses of Rb pathway mechanistic differences among these groups revealed frequent RB1 genomic alterations such as RB1 allelic loss and/or large spanning deletions (83%) in the p16 homo/RB1- subgroups, also indicating that RB1 deletions are frequent in high-grade serous carcinoma. CCNE1 gene gains/amplifications were frequent in the p16 homogeneous/RB1+ subgroup (68%) and cyclin D1 protein overexpression was predominantly characteristic of the p16 heterogeneous/RB1+ subgroup. These subcategories occur early in tumor progression and are seen with similar frequency in the cancer precursor lesion, serous tubal intra-epithelial carcinoma. Overall, this study uniquely identifies multiple non-synonymous mechanisms of retinoblastoma pathway deregulation that correlate with significantly different clinical outcomes. Furthermore, deregulations identified in precursor lesions suggest a key role of this pathway in serous tumor development. Recognition of these categories may identify patients with increased sensitivity to chemotherapy and new opportunities for novel therapeutics.


Assuntos
Cistadenocarcinoma Seroso/metabolismo , Neoplasias Ovarianas/metabolismo , Proteína do Retinoblastoma/metabolismo , Alelos , Biomarcadores Tumorais/metabolismo , Ciclina D1/metabolismo , Ciclina E/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/mortalidade , Intervalo Livre de Doença , Feminino , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , Mutação , Proteínas Oncogênicas/metabolismo , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/mortalidade , Prognóstico , Proteína do Retinoblastoma/genética
3.
J Exp Med ; 210(8): 1529-44, 2013 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-23857982

RESUMO

Oxidative stress plays an important role in cancer development and treatment. Recent data implicate the tumor suppressor BRCA1 in regulating oxidative stress, but the molecular mechanism and the impact in BRCA1-associated tumorigenesis remain unclear. Here, we show that BRCA1 regulates Nrf2-dependent antioxidant signaling by physically interacting with Nrf2 and promoting its stability and activation. BRCA1-deficient mouse primary mammary epithelial cells show low expression of Nrf2-regulated antioxidant enzymes and accumulate reactive oxygen species (ROS) that impair survival in vivo. Increased Nrf2 activation rescues survival and ROS levels in BRCA1-null cells. Interestingly, 53BP1 inactivation, which has been shown to alleviate several defects associated with BRCA1 loss, rescues survival of BRCA1-null cells without restoring ROS levels. We demonstrate that estrogen treatment partially restores Nrf2 levels in the absence of BRCA1. Our data suggest that Nrf2-regulated antioxidant response plays a crucial role in controlling survival downstream of BRCA1 loss. The ability of estrogen to induce Nrf2 posits an involvement of an estrogen-Nrf2 connection in BRCA1 tumor suppression. Lastly, BRCA1-mutated tumors retain a defective antioxidant response that increases the sensitivity to oxidative stress. In conclusion, the role of BRCA1 in regulating Nrf2 activity suggests important implications for both the etiology and treatment of BRCA1-related cancers.


Assuntos
Antioxidantes/metabolismo , Proteína BRCA1/metabolismo , Sobrevivência Celular , Fator 2 Relacionado a NF-E2/metabolismo , Transdução de Sinais , Animais , Proteína BRCA1/deficiência , Proteína BRCA1/genética , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular , Sobrevivência Celular/genética , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Estrogênios/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Inativação de Genes , Humanos , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/patologia , Camundongos , Mutação , NAD(P)H Desidrogenase (Quinona)/genética , NAD(P)H Desidrogenase (Quinona)/metabolismo , Fator 2 Relacionado a NF-E2/genética , Estresse Oxidativo , Ligação Proteica , Estabilidade Proteica , Espécies Reativas de Oxigênio/metabolismo , Ubiquitinação
4.
Sci Transl Med ; 4(142): 142ra95, 2012 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-22786680

RESUMO

Lung cancer is more deadly than colon, breast, and prostate cancers combined, and treatment improvements have failed to improve prognosis significantly. Here, we identify a critical mediator of lung cancer progression, Rac1b, a tumor-associated protein with cell-transforming properties that are linked to the matrix metalloproteinase (MMP)-induced epithelial-mesenchymal transition (EMT) in lung epithelial cells. We show that expression of mouse Rac1b in lung epithelial cells of transgenic mice stimulated EMT and spontaneous tumor development and that activation of EMT by MMP-induced expression of Rac1b gave rise to lung adenocarcinoma in the transgenic mice through bypassing oncogene-induced senescence. Rac1b is expressed abundantly in stages 1 and 2 of human lung adenocarcinomas and, hence, is an attractive molecular target for the development of new therapies that prevent progression to later-stage lung cancers.


Assuntos
Adenocarcinoma/enzimologia , Adenocarcinoma/patologia , Progressão da Doença , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/patologia , Metaloproteinase 3 da Matriz/metabolismo , Neuropeptídeos/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Adenocarcinoma/genética , Adenocarcinoma de Pulmão , Adenoma/enzimologia , Adenoma/genética , Adenoma/patologia , Envelhecimento/patologia , Animais , Linhagem Celular Tumoral , Células Cultivadas , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Mesoderma/metabolismo , Camundongos , Camundongos Transgênicos , Neuropeptídeos/genética , Oncogenes/genética , Vimentina/metabolismo , Proteínas rac de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/genética
5.
Cancer Discov ; 2(9): 826-39, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22777768

RESUMO

UNLABELLED: Although high mammographic density is considered one of the strongest risk factors for invasive breast cancer, the genes involved in modulating this clinical feature are unknown. Tissues of high mammographic density share key histologic features with stromal components within malignant lesions of tumor tissues, specifically low adipocyte and high extracellular matrix (ECM) content. We show that CD36, a transmembrane receptor that coordinately modulates multiple protumorigenic phenotypes, including adipocyte differentiation, angiogenesis, cell-ECM interactions, and immune signaling, is greatly repressed in multiple cell types of disease-free stroma associated with high mammographic density and tumor stroma. Using both in vitro and in vivo assays, we show that CD36 repression is necessary and sufficient to recapitulate the above-mentioned phenotypes observed in high mammographic density and tumor tissues. Consistent with a functional role for this coordinated program in tumorigenesis, we observe that clinical outcomes are strongly associated with CD36 expression. SIGNIFICANCE: CD36 simultaneously controls adipocyte content and matrix accumulation and is coordinately repressed in multiple cell types within tumor and high mammographic density stroma, suggesting that activation of this stromal program is an early event in tumorigenesis. Levels of CD36 and extent of mammographic density are both modifiable factors that provide potential for intervention.


Assuntos
Neoplasias da Mama/diagnóstico por imagem , Antígenos CD36/biossíntese , Células Estromais/metabolismo , Adipócitos/metabolismo , Adipócitos/patologia , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Antígenos CD36/genética , Diferenciação Celular/fisiologia , Feminino , Humanos , Mamografia , Camundongos , Camundongos Knockout , Fatores de Risco , Transdução de Sinais , Células Estromais/patologia
6.
Cancer Prev Res (Phila) ; 4(12): 1953-60, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21920875

RESUMO

p16, a nuclear protein encoded by the p16(INK4a) gene, is a regulator of cell-cycle regulation. Previous studies have shown that expression of p16 in tissue biopsies of patients with ductal carcinoma in situ (DCIS) is associated with increased risk of breast cancer, particularly when considered in combination with other markers such as Ki-67 and COX-2. Here, we evaluated how expression of p16 in breast tissue biopsies of women with atypical hyperplasia (AH), a putative precursor lesion to DCIS, is associated with subsequent development of cancer. p16 expression was assessed by immunohistochemistry in archival sections from 233 women with AH diagnosed at the Mayo Clinic. p16 expression in the atypical lesions was scored by percentage of positive cells and intensity of staining. We also studied coexpression of p16, with Ki-67 and COX-2, biomarkers of progression in AH. Risk factor and follow-up data were obtained via study questionnaire and medical records. Forty-seven patients (20%) developed breast cancer with a median follow-up of 14.5 years. Staining of p16 was increased in older patients relative to younger patients (P = 0.0025). Although risk of developing breast cancer was not associated with increased p16 expression, joint overexpression of Ki-67 and COX-2 was found to convey stronger risk of breast cancer in the first 10 years after diagnosis as compared with one negative marker (P < 0.01). However, the addition of p16 levels did not strengthen this association. p16 overexpression, either alone or in combination with COX-2 and Ki-67, does not significantly stratify breast cancer risk in women with AH.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Hiperplasia/metabolismo , Lesões Pré-Cancerosas/metabolismo , Adulto , Idoso , Neoplasias da Mama/patologia , Ciclo-Oxigenase 2/metabolismo , Feminino , Seguimentos , Humanos , Hiperplasia/patologia , Técnicas Imunoenzimáticas , Antígeno Ki-67/metabolismo , Pessoa de Meia-Idade , Lesões Pré-Cancerosas/patologia , Fatores de Risco
7.
J Natl Cancer Inst ; 102(9): 627-37, 2010 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-20427430

RESUMO

BACKGROUND: Studies have failed to identify characteristics of women who have been diagnosed with ductal carcinoma in situ (DCIS) and have a high or low risk of subsequent invasive cancer. METHODS: We conducted a nested case-control study in a population-based cohort of 1162 women who were diagnosed with DCIS and treated by lumpectomy alone from 1983 to 1994. We collected clinical characteristics and information on subsequent tumors, defined as invasive breast cancer or DCIS diagnosed in the ipsilateral breast containing the initial DCIS lesion or at a regional or distant site greater than 6 months after initial treatment of DCIS (N = 324). We also conducted standardized pathology reviews and immunohistochemical staining for the estrogen receptor (ER), progesterone receptor, Ki67 antigen, p53, p16, epidermal growth factor receptor-2 (ERBB2, HER2/neu oncoprotein), and cyclooxygenase-2 (COX-2) on the initial paraffin-embedded DCIS tissue. Competing risk models were used to determine factors associated with risk of subsequent invasive cancer vs DCIS, and cumulative incidence survival functions were used to estimate 8-year risk. RESULTS: Factors associated with subsequent invasive cancer differed from those associated with subsequent DCIS. Eight-year risk of subsequent invasive cancer was statistically significantly (P = .018) higher for women with initial DCIS lesions that were detected by palpation or that were p16, COX-2, and Ki67 triple positive (p16(+)COX-2(+)Ki67(+)) (19.6%, 95% confidence interval [CI] = 18.0% to 21.3%) than for women with initial lesions that were detected by mammography and were p16, COX-2, and Ki67 triple negative (p16(-)COX-2(-)Ki67(-)) (4.1%, 95% CI = 3.4% to 5.0%). In a multivariable model, DCIS lesions that were p16(+)COX-2(+)Ki67(+) or those detected by palpation were statistically significantly associated with subsequent invasive cancer, but nuclear grade was not. Eight-year risk of subsequent DCIS was highest for women with DCIS lesions that had disease-free margins of 1 mm or greater combined with either ER(-)ERBB2(+)Ki67(+) or p16(+)COX-2(-)Ki67(+) status (23.6%, 95% CI = 18.1% to 34.0%). CONCLUSION: Biomarkers can identify which women who were initially diagnosed with DCIS are at high or low risk of subsequent invasive cancer, whereas histopathology information cannot.


Assuntos
Biomarcadores Tumorais/análise , Neoplasias da Mama/química , Neoplasias da Mama/epidemiologia , Carcinoma Ductal de Mama/diagnóstico , Carcinoma Ductal de Mama/epidemiologia , Carcinoma Intraductal não Infiltrante/química , Mastectomia Segmentar , Recidiva Local de Neoplasia/diagnóstico , Adulto , Idoso , Análise de Variância , Neoplasias da Mama/cirurgia , Carcinoma Intraductal não Infiltrante/cirurgia , Estudos de Casos e Controles , Inibidor p16 de Quinase Dependente de Ciclina , Ciclo-Oxigenase 2/análise , Progressão da Doença , Feminino , Humanos , Imuno-Histoquímica , Incidência , Antígeno Ki-67/análise , Pessoa de Meia-Idade , Proteínas de Neoplasias/análise , Recidiva Local de Neoplasia/epidemiologia , Razão de Chances , Palpação , Valor Preditivo dos Testes , Prognóstico , Receptor ErbB-2/análise , Receptores de Estrogênio/análise , Receptores de Progesterona/análise , Estudos Retrospectivos , Medição de Risco , Fatores de Risco , Análise de Sobrevida , Fatores de Tempo , Proteína Supressora de Tumor p53/análise
8.
Cancer Prev Res (Phila) ; 3(5): 579-87, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20424132

RESUMO

As is well established in invasive breast disease, it is becoming increasingly clear that molecular heterogeneity, both between and within lesions, is a prevalent, distinct phenotype of premalignant lesions of the breast. Key pathways of tumorigenesis modulate critical features of premalignant lesions such as proliferation, differentiation, stress response, and even the generation of diversity. Current studies show that evaluation of these lesions may provide clinically useful information on future tumor formation as well as biological insights into the origin and functional significance of this distinct phenotype.


Assuntos
Neoplasias da Mama/patologia , Lesões Pré-Cancerosas/patologia , Animais , Neoplasias da Mama/genética , Feminino , Humanos , Camundongos , Lesões Pré-Cancerosas/genética , Fatores de Risco
9.
Breast Cancer Res ; 11(6): R87, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19995452

RESUMO

INTRODUCTION: We have previously identified a rare subpopulation of variant human mammary epithelial cells (vHMEC) with repressed p16INK4A that exist in disease-free women yet display premalignant properties, suggesting that they have engaged the process of malignant transformation. In order to gain insight into the molecular alterations required for vHMEC to progress to malignancy, and to characterize the epigenetic events associated with early progression, we examined the effect of oncogenic stress on the behavior of these cells. METHODS: HMEC that express p16INK4A and vHMEC that do not, were transduced with constitutively active Ha-rasV12 and subsequently exposed to serum to determine whether signals from the cellular microenvironment could cooperate with ras to promote the malignant transformation of vHMEC. Epigenetic alterations were assessed using methylation-specific polymerase chain reaction (PCR). RESULTS: vHMEC expressing Ha-rasV12 (vHMEC-ras) bypassed the classic proliferative arrest that has been previously documented in normal fibroblasts following oncogenic stress, and that we also observe here in normal HMEC. Moreover, vHMEC-ras cells exhibited many additional alterations that are observed during progression to malignancy such as the generation of chromosomal abnormalities, upregulation of telomerase activity, immortalization following exposure to serum, and anchorage-independent growth, but they did not form tumors following orthotopic injection in vivo. Associated with their early progression to malignancy was an increase in the number of genes methylated, two of which (RASSF1A and SFRP1) were also methylated in other immortalized mammary cell lines as well as in breast cancer cells and tissues. CONCLUSIONS: We have characterized a mammary progression model that recapitulates molecular and methylation alterations observed in many breast cancers. Our data suggest that concomitant methylation of RASSF1A and SFRP1 marks an early event in mammary transformation and may thus have prognostic potential.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Metilação de DNA , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/patologia , Animais , Adesão Celular/fisiologia , Processos de Crescimento Celular/genética , Linhagem Celular Tumoral , Aberrações Cromossômicas , Progressão da Doença , Feminino , Genes p16 , Genes ras , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteínas de Membrana/genética , Camundongos , Camundongos SCID , Telomerase/metabolismo , Proteínas Supressoras de Tumor/genética , Regulação para Cima
10.
Cancer Cell ; 12(5): 479-91, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17996651

RESUMO

Approximately 15%-30% of women diagnosed with ductal carcinoma in situ (DCIS) develop a subsequent tumor event within 10 years after surgical lumpectomy. To date, little is known about the molecular pathways that confer this differential risk for developing subsequent disease. In this study, we demonstrate that expression of biomarkers indicative of an abrogated response to cellular stress predicts DCIS with worse outcome and is a defining characteristic of basal-like invasive tumors. Mechanistic studies identify the Rb pathway as a key regulator of this response. Conversely, biomarkers indicative of an intact response to cellular stress are strongly associated with a disease-free prognosis. Assessment of these biomarkers in DCIS begins to allow prediction of tumor formation years before it actually occurs.


Assuntos
Biomarcadores Tumorais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carcinoma Intraductal não Infiltrante/metabolismo , Carcinoma Intraductal não Infiltrante/patologia , Ciclo Celular , Proliferação de Células , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Ciclo-Oxigenase 2/metabolismo , Feminino , Humanos , Modelos Biológicos , Prognóstico , RNA Mensageiro/metabolismo , Proteína do Retinoblastoma/metabolismo , Risco , Resultado do Tratamento
11.
Cancer Res ; 66(21): 10325-31, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17079452

RESUMO

p16(INK4a) (p16) and p53 are tumor suppressor genes that are inactivated during carcinogenesis in many tumors. Here we show that p16 gene activity inversely modulates p53 status and function in primary human mammary epithelial cells. Reduced levels of p16 protein stabilize p53 protein through inhibition of proteolytic degradation, and this increase in p53 protein levels enhances the cellular response to radiation, represses proliferation, and transcriptionally activates downstream targets. Stabilization of p53 is mediated through the retinoblastoma/E2F/p14(ARF)/murine double minute-2 pathway. However, we have observed that p16 does not modulate p53 in fibroblasts, indicating a possible cell type-specific regulation of this pathway.


Assuntos
Neoplasias da Mama/etiologia , Mama/citologia , Inibidor p16 de Quinase Dependente de Ciclina/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Proliferação de Células , Células Cultivadas , Inibidor p16 de Quinase Dependente de Ciclina/análise , Células Epiteliais/citologia , Feminino , Genes p16 , Humanos , Proteína do Retinoblastoma/fisiologia , Transdução de Sinais , Proteína Supressora de Tumor p53/análise
12.
Cancer Res ; 65(5): 1792-9, 2005 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-15753376

RESUMO

The immediate-early gene, cyclooxygenase-2 (COX-2), is induced in a variety of inflammatory and neoplastic processes and is believed to play an important role in tumorigenesis. In this study, we identify an important upstream regulatory pathway of COX-2 expression in variant human mammary epithelial cells (vHMEC), which has been shown to exhibit phenotypes important for malignancy. We find that the stress-activated kinase, p38, is phosphorylated and activated in vHMEC compared with HMEC and is responsible for the expression of COX-2 in vHMEC as cells grow in culture. Furthermore in this capacity, p38 acts to stabilize the COX-2 transcript rather than activate COX-2 transcription. Inhibition of p38 kinase, using a chemical inhibitor, down-regulates COX-2 and decreases cell survival. Examination of archived tissue from women with ductal carcinoma in situ reveals epithelial cells that not only overexpress COX-2 but also have an abundance of activated phospho-p38 in the nucleus and cytoplasm, mirroring the expression observed in vitro. These epithelial cells are found within premalignant lesions as well as in fields of morphologically normal tissue that surround the lesions. In contrast, low phospho-p38 staining was observed in the majority of normal tissue obtained from reduction mammoplasty. These data help define the regulation of COX-2 expression in early carcinogenesis and provide alternative candidates for targeted prevention of COX-2-induced phenotypes and breast cancer.


Assuntos
Neoplasias da Mama/enzimologia , Carcinoma Intraductal não Infiltrante/enzimologia , Células Epiteliais/enzimologia , Regulação Enzimológica da Expressão Gênica , Prostaglandina-Endoperóxido Sintases/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Apoptose/efeitos dos fármacos , Mama/enzimologia , Mama/patologia , Neoplasias da Mama/patologia , Carcinoma Intraductal não Infiltrante/patologia , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Ciclo-Oxigenase 2 , Citoplasma/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Proteínas de Membrana , Pessoa de Meia-Idade , Fosforilação , Lesões Pré-Cancerosas/enzimologia , Lesões Pré-Cancerosas/patologia , Prostaglandina-Endoperóxido Sintases/genética , Prostaglandinas/metabolismo , Transcrição Gênica , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/genética
13.
J Mammary Gland Biol Neoplasia ; 9(3): 263-74, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15557799

RESUMO

Studies of human mammary epithelial cells from healthy individuals are providing novel insights into how early epigenetic and genetic events affect genomic integrity and fuel carcinogenesis. Key epigenetic changes, such as the hypermethylation of the p16 (INK4a) promoter sequences, create a previously unappreciated preclonal phase of tumorigenesis in which a subpopulation of mammary epithelial cells are positioned for progression to malignancy (Romanov et al. , 2001, Nature , 409:633-637; Tlsty et al. , 2001, J. Mammary Gland Biol. Neoplasia , 6:235-243). These key changes precede the clonal outgrowth of premalignant lesions and occur frequently in healthy, disease-free women. Understanding more about these early events should provide novel molecular candidates for prevention and therapy of breast cancer that target the process instead of the consequences of genomic instability. This review will highlight some of the key alterations that have been studied in human mammary epithelial cells in culture and relate them to events observed in vivo and discussed in accompanying reviews in this volume.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/fisiopatologia , Transformação Celular Neoplásica/genética , Regulação Neoplásica da Expressão Gênica , Glândulas Mamárias Humanas/citologia , Epigênese Genética , Células Epiteliais/fisiologia , Feminino , Fibroblastos/fisiologia , Perfilação da Expressão Gênica , Instabilidade Genômica , Humanos
14.
Cancer Cell ; 5(3): 263-73, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15050918

RESUMO

Breast tissue from healthy women contains variant mammary epithelial cells (vHMEC) exhibiting p16INK4a promoter hypermethylation both in vivo and in vitro. When continuously cultured, vHMEC acquire telomeric dysfunction and produce the types of chromosomal abnormalities seen in premalignant lesions of cancer. We find that late passage vHMEC express elevated prostaglandin cyclo-oxygenase 2 (COX-2), which contributes to increased prostaglandin synthesis, angiogenic activity, and invasive ability. These data demonstrate the existence of human mammary epithelial cells with the potential to acquire multiple genomic alterations and phenotypes associated with malignant cells. Moreover, COX-2 overexpression coincides with focal areas of p16INK4a hypermethylation in vivo, creating ideal candidates as precursors to breast cancer. These putative precursors can be selectively eliminated upon exposure to COX-2 inhibitors in vitro.


Assuntos
Mama/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Células Epiteliais/metabolismo , Isoenzimas/metabolismo , Lesões Pré-Cancerosas/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Apoptose/fisiologia , Mama/fisiopatologia , Aberrações Cromossômicas , Ciclo-Oxigenase 2 , Dano ao DNA/fisiologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Proteínas de Membrana , Metilação , Neovascularização Patológica/metabolismo , Prostaglandinas/metabolismo , Telomerase/metabolismo
15.
Biochem Biophys Res Commun ; 307(4): 839-46, 2003 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-12878187

RESUMO

Previous work has shown that phorbol esters modulate chemotaxis. Here, we demonstrate that PKC activation via phorbol 12-myristate 13-acetate (PMA) treatment of MDA-MB-231 cells inhibits EGF-induced cell spreading, the initial event of motility and chemotaxis. Of five PKC isoforms (alpha,iota,lambda,delta,and epsilon) identified in this cell line, PMA treatment only induced PKCalpha translocation from the cytosol to the membrane, an event that correlated with the development of the rounded morphology. Cell recovery was linked to PKCalpha downregulation in part via the proteasome pathway since treatment with MG101 in the presence of PMA did not lead to PKCalpha degradation and cell recovery. Co-immunoprecipitation and immunolocalization demonstrated that EGF co-localized with PKCalpha and EGFR, however, PMA did not abrogate EGFR transactivation. This work suggests that PKCalpha is the primary target of PMA acting as a transient negative regulator of cell spreading and motility in MDA-MB-231 breast cancer cells.


Assuntos
Neoplasias da Mama/enzimologia , Movimento Celular , Receptores ErbB/metabolismo , Proteína Quinase C/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Tamanho Celular , Fator de Crescimento Epidérmico/análise , Fator de Crescimento Epidérmico/antagonistas & inibidores , Receptores ErbB/análise , Feminino , Humanos , Proteína Quinase C/análise , Proteína Quinase C-alfa , Transdução de Sinais , Acetato de Tetradecanoilforbol/farmacologia , Células Tumorais Cultivadas
16.
Cancer Res ; 63(10): 2347-50, 2003 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-12750248

RESUMO

Cyclooxygenase-2 (COX-2) is emerging as an important cancer biomarker and is now an experimental target for solid tumor treatment.However, no study has exclusively focused on COX-2 expression in early lesions such as ductal carcinoma in situ (DCIS). We examined COX-2 expression by immunohistochemistry in 46 cases of women undergoing surgical resection for DCIS. We found that COX-2 expression was detected in 85% of all DCIS specimens, with increased COX-2 staining correlating with higher nuclear grade. Strikingly, COX-2 staining intensity in the normal adjacent epithelium was stronger than in the DCIS lesion itself. Our observations demonstrate that COX-2 is up-regulated in the normal adjacent epithelium and supports the hypothesis that the surrounding epithelial tissue is part of the disease process in DCIS.


Assuntos
Neoplasias da Mama/enzimologia , Carcinoma in Situ/enzimologia , Carcinoma Ductal de Mama/enzimologia , Isoenzimas/biossíntese , Prostaglandina-Endoperóxido Sintases/biossíntese , Adulto , Idoso , Neoplasias da Mama/patologia , Carcinoma in Situ/patologia , Carcinoma Ductal de Mama/patologia , Núcleo Celular/patologia , Ciclo-Oxigenase 2 , Células Epiteliais/enzimologia , Feminino , Humanos , Imuno-Histoquímica , Proteínas de Membrana , Pessoa de Meia-Idade , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA