Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Breast Cancer Res ; 22(1): 122, 2020 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-33148314

RESUMO

BACKGROUND: The role of nuclear receptors in both the aetiology and treatment of breast cancer is exemplified by the use of the oestrogen receptor (ER) as a prognostic marker and treatment target. Treatments targeting the oestrogen signalling pathway are initially highly effective for most patients. However, for the breast cancers that fail to respond, or become resistant, to current endocrine treatments, the long-term outlook is poor. ER is a member of the nuclear receptor superfamily, comprising 48 members in the human, many of which are expressed in the breast and could be used as alternative targets in cases where current treatments are ineffective. METHODS: We used sparse canonical correlation analysis to interrogate potential novel nuclear receptor expression relationships in normal breast and breast cancer. These were further explored using whole transcriptome profiling in breast cancer cells after combinations of ligand treatments. RESULTS: Using this approach, we discovered a tumour suppressive relationship between the mineralocorticoid receptor (MR) and retinoic acid receptors (RAR), in particular RARß. Expression profiling of MR expressing breast cancer cells revealed that mineralocorticoid and retinoid co-treatment activated an expression program consistent with a reverse Warburg effect and growth inhibition, which was not observed with either ligand alone. Moreover, high expression of both MR and RARB was associated with improved breast cancer-specific survival. CONCLUSION: Our study reveals a previously unknown relationship between MR and RAR in the breast, which is dependent on menopausal state and altered in malignancy. This finding identifies potential new targets for the treatment of breast cancers that are refractory to existing therapeutic options.


Assuntos
Neoplasias da Mama/patologia , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Receptores de Mineralocorticoides/metabolismo , Receptores do Ácido Retinoico/metabolismo , Transdução de Sinais , Efeito Warburg em Oncologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Biologia Computacional , Feminino , Humanos , Receptores de Estrogênio/metabolismo , Receptores de Mineralocorticoides/genética , Taxa de Sobrevida , Células Tumorais Cultivadas
2.
J Mammary Gland Biol Neoplasia ; 20(1-2): 27-37, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26254191

RESUMO

The epithelium of the human breast is made up of a branching ductal-lobular system, which is lined by a single layer of luminal cells surrounded by a contractile basal cell layer. The co-ordinated development of stem/progenitor cells into these luminal and basal cells is fundamentally important for breast morphogenesis. The ovarian steroid hormone, progesterone, is critical in driving proliferation and normal breast development, yet progesterone analogues have also been shown to be a major driver of breast cancer risk. Studies in recent years have revealed an important role for progesterone in stimulating the mammary stem cell compartment in the mouse mammary gland, and growing evidence supports the notion that progesterone also stimulates progenitor cells in both the normal human breast and in breast cancer cells. As changes in cell type composition are one of the hallmark features of breast cancer progression, these observations have critical implications in discerning the mechanisms of how progesterone increases breast cancer risk. This review summarises recent work regarding the impact of progesterone action on the stem/progenitor cell compartment of the human breast.


Assuntos
Neoplasias da Mama/metabolismo , Mama/metabolismo , Epitélio/metabolismo , Progesterona/metabolismo , Células-Tronco/metabolismo , Comunicação Autócrina , Mama/crescimento & desenvolvimento , Linhagem Celular Tumoral , Feminino , Humanos , Glândulas Mamárias Humanas/metabolismo , Receptores de Progesterona/metabolismo , Transdução de Sinais
3.
Breast Cancer Res Treat ; 151(2): 309-18, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25917868

RESUMO

Progesterone receptor (PR) function, while essential in normal human breast, is also implicated in breast cancer risk. The two progesterone receptors, PRA and PRB, are co-expressed at equivalent levels in normal breast, but early in carcinogenesis normal levels of PRA:PRB are frequently disrupted, and predominance of one isoform, usually PRA, results. In model systems, PRA and PRB have different activities, and altering the PRA:PRB ratio in cell lines alters PR signaling. The purpose of this study was to determine whether hormonal or reproductive factors contribute to imbalanced PRA:PRB expression in breast tumors and the impact of PRA:PRB imbalance on disease outcome. The relative expression of PRA and PRB proteins was determined by dual immunofluorescence histochemistry in archival breast tumors and associations with clinical and reproductive history assessed. PRA:PRB expression was not influenced by reproductive factors, whereas exogenous hormone use (menopausal hormone treatment, MHT) favored PRB expression (p < 0.035). The PRA:PRB ratio may be a discriminator of response to endocrine therapy in the TransATAC sample collection, with high PRA:PRB ratio predicting earlier relapse for women on tamoxifen, but not anastrozole (mean lnPRA:PRB ratio; HR (95 % CI) tamoxifen 2.45 (1.20-4.99); p value 0.02; anastrozole 0.80 (0.36-1.78); p value 0.60). The results of this study show that PRA:PRB imbalance in breast cancers is not associated with lifetime endogenous endocrine and reproductive factors, but is associated with MHT use, and that PRA predominance can discriminate those women who will relapse earlier on tamoxifen treatment. These data support a role for imbalanced PRA:PRB expression in breast cancer progression and relative benefit from endocrine treatment.


Assuntos
Antineoplásicos Hormonais/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Receptores de Progesterona/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Estudos de Coortes , Feminino , Expressão Gênica , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , Gradação de Tumores , Metástase Neoplásica , Isoformas de Proteínas , Receptores de Progesterona/genética , Fatores de Risco , Resultado do Tratamento , Carga Tumoral
4.
Breast Cancer Res ; 16(3): 102, 2014 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-25928365

RESUMO

A key approach in understanding how breast cancer can occur is to determine the regulatory pathways at play in the normal breast and to identify precisely the normal developmental mechanisms subverted during early breast cancer progression. Using normal human breast tissue samples, Pardo and colleagues have identified the gene targets and pathways displaying fluctuating expression as a consequence of the menstrual cycle. Detailed characterization of how the human breast functions in its normal state, and how this may be perturbed at its earliest point, will provide a critical step toward the prevention of breast cancer.


Assuntos
Mama/metabolismo , Epitélio/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Pré-Menopausa/genética , Bancos de Tecidos , Transcriptoma/genética , Feminino , Humanos
5.
Breast Cancer Res Treat ; 143(3): 423-33, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24395108

RESUMO

The epithelium of the human breast is made up of a branching ductal-lobular system, which is lined by a single layer of luminal cells surrounded by a contractile basal cell layer. The co-ordinated development of stem/progenitor cells into these luminal and basal cells is fundamentally important for breast morphogenesis. The ovarian steroid hormones, progesterone (P) and 17ß-estradiol, are critical in driving this normal breast development, yet ovarian activity has also been shown to be a major driver of breast cancer risk. We previously demonstrated that P treatment increases proliferation and augments the number of progenitor-like cells, and that the progesterone receptor (PR) is also expressed in the bipotent progenitor-enriched subfraction. Here we demonstrate that PR is expressed in a subset of CD10+ basal cells and that P stimulates this CD10+ cell compartment, which is enriched for bipotent progenitor activity. In addition, we have shown that P stimulates progenitor cells in human breast cancer cell lines and expands the cancer stem cell population via increasing the stem-like CD44+ population. As changes in cell type composition are one of the hallmark features of breast cancer progression, the demonstration that progenitor cells are stimulated by P in both normal breast and in breast cancer cells has critical implications in discerning the mechanisms of how P increases breast cancer risk.


Assuntos
Neoplasias da Mama/metabolismo , Mama/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Progesterona/administração & dosagem , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Linhagem da Célula , Estradiol/administração & dosagem , Estradiol/genética , Feminino , Humanos , Receptores de Hialuronatos/genética , Células-Tronco Neoplásicas/efeitos dos fármacos , Neprilisina/genética , Progesterona/genética , Receptores de Estrogênio/metabolismo , Células-Tronco/efeitos dos fármacos
7.
Biochim Biophys Acta ; 1783(3): 383-93, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18190796

RESUMO

Mammary gland development is coupled to reproductive events by hormonal cues of ovarian and pituitary origin, which activate a genomic regulatory network. Identification of the components and regulatory links that comprise this network will provide the basis for defining the network's dynamic response during normal development and its perturbation during breast carcinogenesis. In this study KIBRA was identified as a transcript showing decreased expression associated with failed mammary gland development in Prlr knockout mammary epithelium. It is strongly up-regulated during pregnancy, falls during lactation and is again up-regulated during involution of the gland at weaning. A bioinformatic approach was undertaken to identify potential binding partners which interact with the WW domains of KIBRA. We show that KIBRA binds to a WW domain binding motif, PPxY, in the tyrosine kinase receptor DDR1, and dissociates upon treatment with the DDR1 ligands collagen type I or IV. In addition we show that KIBRA and DDR1 also interact with PKCz to form a trimeric complex. Finally, overexpression and knockdown studies demonstrate that KIBRA promotes the collagen-stimulated activation of the MAPK cascade. Thus KIBRA may play a role in how the reproductive state influences the mammary epithelial cell to respond to changing cell-context information, such as experienced during the tissue remodeling events of mammary gland development.


Assuntos
Colágeno/fisiologia , Proteínas/metabolismo , Proteínas/fisiologia , Receptores Proteína Tirosina Quinases/metabolismo , Receptores Mitogênicos/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Receptores com Domínio Discoidina , Ativação Enzimática/fisiologia , Feminino , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Sistema de Sinalização das MAP Quinases/fisiologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Glândulas Mamárias Humanas/metabolismo , Glândulas Mamárias Humanas/fisiologia , Camundongos , Camundongos Knockout , Morfogênese/genética , Fosfoproteínas , Gravidez , Ligação Proteica , Receptores Proteína Tirosina Quinases/genética , Receptores Mitogênicos/genética , Receptores da Prolactina/genética , Transdução de Sinais/fisiologia
8.
Mol Cell Endocrinol ; 466: 2-14, 2018 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-28851667

RESUMO

The ovarian hormones estrogen and progesterone are master regulators of the development and function of a broad spectrum of human tissues, including the breast, reproductive and cardiovascular systems, brain and bone. Acting through the nuclear estrogen (ER) and progesterone receptors (PR), both play complex and essential coordinated roles in the extensive development of the lobular alveolar epithelial structures of the normal breast during puberty, the normal menstrual cycle and pregnancy. The past decade has seen major advances in understanding the mechanisms of action of estrogen and progesterone in the normal breast and in the delineation of the complex hierarchy of cell types regulated by ovarian hormones in this tissue. There is evidence for a role for both ER and PR in driving breast cancer, and both are favourable prognostic markers with respect to outcome. In this review, we summarize current knowledge of the mechanisms of action of ER and PR in the normal breast, and implications for the development and management of breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Mama , Estrogênios/metabolismo , Progesterona/metabolismo , Animais , Mama/metabolismo , Mama/patologia , Neoplasias da Mama/patologia , Feminino , Humanos , Camundongos , Camundongos Knockout , Receptores de Estrogênio/química , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/química , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo
9.
Mol Endocrinol ; 20(5): 1177-87, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16469767

RESUMO

The proliferative phase of mammary alveolar morphogenesis is initiated during early pregnancy by rising levels of serum prolactin and progesterone, establishing a program of gene expression that is ultimately responsible for the development of the lobuloalveoli and the onset of lactation. To explore this largely unknown genetic program, we constructed transcript profiles derived from transplanted mammary glands formed by recombination of prolactin receptor (Prlr) knockout or wild-type mammary epithelium with wild-type mammary stroma. Comparison with profiles derived from prolactin-treated Scp2 mammary epithelial cells produced a small set of commonly prolactin-regulated genes that included the negative regulator of cytokine signaling, Socs2 (suppressor of cytokine signaling 2), and the ets transcription factor, E74-like factor 5 (Elf5). Homozygous null mutation of Socs2 rescued the failure of lactation and reduction of mammary signal transducer and activator of transcription 5 phosphorylation that characterizes Prlr heterozygous mice, demonstrating that mammary Socs2 is a key regulator of the prolactin-signaling pathway. Reexpression of Elf5 in Prlr nullizygous mammary epithelium restored lobuloalveolar development and milk production, demonstrating that Elf5 is a transcription factor capable of substituting for prolactin signaling. Thus, Socs2 and Elf5 are key members of the set of prolactin-regulated genes that mediate prolactin-driven mammary development.


Assuntos
Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica no Desenvolvimento , Glândulas Mamárias Animais/crescimento & desenvolvimento , Prolactina/fisiologia , Proteínas Supressoras da Sinalização de Citocina/genética , Fatores de Transcrição/genética , Animais , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/metabolismo , Feminino , Perfilação da Expressão Gênica , Lactação/genética , Glândulas Mamárias Animais/química , Glândulas Mamárias Animais/efeitos dos fármacos , Camundongos , Camundongos Knockout , Gravidez , Prolactina/farmacologia , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Receptores da Prolactina/genética , Proteínas Supressoras da Sinalização de Citocina/análise , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Fatores de Transcrição/análise , Fatores de Transcrição/metabolismo , Transcrição Gênica
10.
Breast Cancer Res ; 8(2): 207, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16677418

RESUMO

Massive tissue remodelling occurs within the mammary gland during pregnancy, resulting in the formation of lobuloalveoli that are capable of milk secretion. Endocrine signals generated predominantly by prolactin and progesterone operate the alveolar switch to initiate these developmental events. Here we review the current understanding of the components of the alveolar switch and conclude with an examination of the role of the ets transcription factor Elf5. We propose that Elf5 is a key regulator of the alveolar switch.


Assuntos
Mama/fisiologia , Células Epiteliais/fisiologia , Glândulas Mamárias Humanas/fisiologia , Gravidez/fisiologia , Mama/citologia , Mama/crescimento & desenvolvimento , Divisão Celular , Células Epiteliais/citologia , Feminino , Humanos , Glândulas Mamárias Humanas/citologia , Glândulas Mamárias Humanas/crescimento & desenvolvimento , Morfogênese/fisiologia , Progesterona/fisiologia , Prolactina/fisiologia , Fatores de Transcrição/metabolismo , beta Catenina/fisiologia
11.
Mol Endocrinol ; 29(9): 1230-42, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26266959

RESUMO

Progesterone (P), which signals through the P receptor (PR), is critical in normal development of the breast, but its signaling axis is also a major driver of breast cancer risk. Here we review recent advances in the understanding of P signaling in the normal human breast, with a focus on the importance of the balance between autocrine and paracrine signaling. To date, most data (which derive largely from mouse models or human breast cancer cell line studies) have demonstrated that the vast majority of PR+ cells appear to act as "sensor" cells, which respond to P stimulation by translating these hormonal cues into paracrine signals. However, growing evidence suggests that, dependent on the cellular context, P may also signal in an autocrine manner in a subset of cells in the normal mouse mammary gland and human breast. It has been suggested that it may be dysregulation of this autocrine signaling, resulting in a "switch" from a predominance of paracrine signaling to autocrine signaling in PR+ cells, which is an early event during breast tumorigenesis. This review summarizes current evidence in the literature that demonstrates the mechanisms through which P acts in the normal human breast, as well as highlighting the important questions that remain unanswered.


Assuntos
Comunicação Autócrina/fisiologia , Neoplasias da Mama/patologia , Comunicação Parácrina/fisiologia , Progesterona/metabolismo , Receptores de Progesterona/metabolismo , Animais , Mama/metabolismo , Proliferação de Células/fisiologia , Modelos Animais de Doenças , Feminino , Humanos , Glândulas Mamárias Animais/metabolismo , Camundongos , Transdução de Sinais/fisiologia
12.
Oncotarget ; 5(18): 8651-64, 2014 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-25261374

RESUMO

Cumulative exposure to estrogen (E) and progesterone (P) over the menstrual cycle significantly influences the risk of developing breast cancer. Despite the dogma that PR in the breast merely serves as a marker of an active estrogen receptor (ER), and as an inhibitor of the proliferative actions of E, it is now clear that in the breast P increases proliferation independently of E action. We show here that the progesterone receptor (PR) and ER are expressed in different epithelial populations, and target non-overlapping pathways in the normal human breast. In breast cancer, PR becomes highly correlated with ER, and this convergence is associated with signaling pathways predictive of disease metastasis. These data challenge the established paradigm that ER and PR function co-operatively in normal breast, and have significant implications not only for our understanding of normal breast biology, but also for diagnosis, prognosis and/or treatment options in breast cancer patients.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/metabolismo , Carcinoma Intraductal não Infiltrante/metabolismo , Transformação Celular Neoplásica/metabolismo , Células Epiteliais/metabolismo , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Humanas/metabolismo , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Transdução de Sinais , Animais , Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Carcinoma Intraductal não Infiltrante/genética , Carcinoma Intraductal não Infiltrante/mortalidade , Carcinoma Intraductal não Infiltrante/secundário , Estudos de Casos e Controles , Linhagem da Célula , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Células Epiteliais/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Estimativa de Kaplan-Meier , Glândulas Mamárias Humanas/patologia , Prognóstico , RNA Mensageiro/metabolismo , Receptor Cross-Talk , Receptores de Estrogênio/genética , Receptores de Progesterona/genética , Fatores de Tempo
13.
Histol Histopathol ; 28(9): 1197-204, 2013 09.
Artigo em Inglês | MEDLINE | ID: mdl-23584793

RESUMO

The epithelium compartment of the human breast is made up of a branching ductal-lobular system, which is lined by a single layer of luminal epithelial cells surrounded by contractile myoepithelial cells. The co-ordinated development of these two cell types, and maintenance of their relative proportions, is fundamentally important for normal breast morphogenesis. Changes in cell type composition is one of the hallmark features of breast cancer progression, and the vast majority of breast tumors are comprised of luminal cells only, with a complete absence of myoepithelial cells. Despite this striking alteration in relative proportions of luminal and myoepithelial cells in invasive breast cancers compared with normal breast tissue, the steps in this dramatic change in cellular composition remain poorly characterised, nor is it known whether loss of myoepithelial cells is an early event in carcinogenesis. In a panel of breast tissues, we quantitated the proportion of luminal cells relative to the surrounding myoepithelial cell layer in a panel of normal and pre-invasive breast tissue samples, including lesions with proliferative disease without atypia (PDWA), columnar cell lesions (CCL), atypical ductal hyperplasia (ADH), and DCIS, and correlated these findings with proliferation in the same lesions. The study findings showed that changes in lineage composition correlate with increased proliferation, and are one of the earliest events in breast carcinogenesis. Therefore not only are myoepithelial cells important in distinguishing between invasive and non-invasive tumors, their relative proportion compared with luminal cell numbers may provide a new potential indicator of which premalignant lesions are at higher risk of progression to invasive disease.


Assuntos
Neoplasias da Mama/patologia , Carcinogênese , Linhagem da Célula , Regulação Neoplásica da Expressão Gênica , Carcinoma Intraductal não Infiltrante/patologia , Proliferação de Células , Transformação Celular Neoplásica , Epitélio/patologia , Feminino , Humanos , Hiperplasia/metabolismo , Microscopia de Fluorescência , Prognóstico , Fatores de Risco
14.
Genes Dev ; 22(5): 581-6, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18316476

RESUMO

Hormonal cues regulate mammary development, but the consequent transcriptional changes and cell fate decisions are largely undefined. We show that knockout of the prolactin-regulated Ets transcription factor Elf5 prevented formation of the secretory epithelium during pregnancy. Conversely, overexpression of Elf5 in an inducible transgenic model caused alveolar differentiation and milk secretion in virgin mice, disrupting ductal morphogenesis. CD61+ luminal progenitor cells accumulated in Elf5-deficient mammary glands and were diminished in glands with Elf5 overexpression. Thus Elf5 specifies the differentiation of CD61+ progenitors to establish the secretory alveolar lineage during pregnancy, providing a link between prolactin, transcriptional events, and alveolar development.


Assuntos
Diferenciação Celular/genética , Proteínas de Ligação a DNA/metabolismo , Lactação/genética , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/metabolismo , Morfogênese/genética , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo , Animais , Linhagem da Célula/genética , Proteínas de Ligação a DNA/genética , Epitélio/crescimento & desenvolvimento , Epitélio/metabolismo , Feminino , Integrina beta3/análise , Glândulas Mamárias Animais/citologia , Camundongos , Camundongos Transgênicos , Gravidez , Células-Tronco/citologia , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA