Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Development ; 140(1): 117-25, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23154416

RESUMO

Tyrosine phosphorylation plays a fundamental role in mammary gland development. However, the role of specific tyrosine phosphatases in controlling mammary cell fate remains ill defined. We have identified protein tyrosine phosphatase 1B (PTP1B) as an essential regulator of alveologenesis and lactogenesis. PTP1B depletion increased the number of luminal mammary progenitors in nulliparous mice, leading to enhanced alveoli formation upon pregnancy. Mechanistically, Ptp1b deletion enhanced the expression of progesterone receptor and phosphorylation of Stat5, two key regulators of alveologenesis. Furthermore, glands from Ptp1b knockout mice exhibited increased expression of milk proteins during pregnancy due to enhanced Stat5 activation. These findings reveal that PTP1B constrains the number of mammary progenitors and thus prevents inappropriate onset of alveologenesis in early pregnancy. Moreover, PTP1B restrains the expression of milk proteins during pregnancy and thus prevents premature lactogenesis. Our work has implications for breast tumorigenesis because Ptp1b deletion has been shown to prevent or delay the onset of mammary tumors.


Assuntos
Diferenciação Celular/fisiologia , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/enzimologia , Proteína Tirosina Fosfatase não Receptora Tipo 1/fisiologia , Células-Tronco/metabolismo , Animais , Diferenciação Celular/genética , Células Cultivadas , Feminino , Lactação/genética , Masculino , Glândulas Mamárias Animais/embriologia , Camundongos , Camundongos Knockout , Gravidez , Progesterona/antagonistas & inibidores , Progesterona/biossíntese , Progesterona/fisiologia , Proteína Tirosina Fosfatase não Receptora Tipo 1/deficiência , Proteína Tirosina Fosfatase não Receptora Tipo 1/genética , Fator de Transcrição STAT5/antagonistas & inibidores , Fator de Transcrição STAT5/biossíntese , Fator de Transcrição STAT5/fisiologia , Células-Tronco/citologia , Células-Tronco/enzimologia , Regulação para Cima/genética
2.
Proc Natl Acad Sci U S A ; 109(8): 2772-7, 2012 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-21940501

RESUMO

Human breast cancers are broadly classified based on their gene-expression profiles into luminal- and basal-type tumors. These two major tumor subtypes express markers corresponding to the major differentiation states of epithelial cells in the breast: luminal (EpCAM(+)) and basal/myoepithelial (CD10(+)). However, there are also rare types of breast cancers, such as metaplastic carcinomas, where tumor cells exhibit features of alternate cell types that no longer resemble breast epithelium. Until now, it has been difficult to identify the cell type(s) in the human breast that gives rise to these various forms of breast cancer. Here we report that transformation of EpCAM(+) epithelial cells results in the formation of common forms of human breast cancer, including estrogen receptor-positive and estrogen receptor-negative tumors with luminal and basal-like characteristics, respectively, whereas transformation of CD10(+) cells results in the development of rare metaplastic tumors reminiscent of the claudin-low subtype. We also demonstrate the existence of CD10(+) breast cells with metaplastic traits that can give rise to skin and epidermal tissues. Furthermore, we show that the development of metaplastic breast cancer is attributable, in part, to the transformation of these metaplastic breast epithelial cells. These findings identify normal cellular precursors to human breast cancers and reveal the existence of a population of cells with epidermal progenitor activity within adult human breast tissues.


Assuntos
Neoplasias da Mama/patologia , Transformação Celular Neoplásica/patologia , Adulto , Antígenos de Neoplasias/metabolismo , Neoplasias da Mama/metabolismo , Moléculas de Adesão Celular/metabolismo , Transformação Celular Neoplásica/metabolismo , Molécula de Adesão da Célula Epitelial , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Epitélio/metabolismo , Epitélio/patologia , Feminino , Humanos , Metaplasia , Neprilisina/metabolismo , Fenótipo
3.
Breast Cancer Res ; 15(2): R36, 2013 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-23621987

RESUMO

INTRODUCTION: Early pregnancy has a strong protective effect against breast cancer in humans and rodents, but the underlying mechanism is unknown. Because breast cancers are thought to arise from specific cell subpopulations of mammary epithelia, we studied the effect of parity on the transcriptome and the differentiation/proliferation potential of specific luminal and basal mammary cells in mice. METHODS: Mammary epithelial cell subpopulations (luminal Sca1-, luminal Sca1+, basal stem/progenitor, and basal myoepithelial cells) were isolated by flow cytometry from parous and age-matched virgin mice and examined by using a combination of unbiased genomics, bioinformatics, in vitro colony formation, and in vivo limiting dilution transplantation assays. Specific findings were further investigated with immunohistochemistry in entire glands of parous and age-matched virgin mice. RESULTS: Transcriptome analysis revealed an upregulation of differentiation genes and a marked decrease in the Wnt/Notch signaling ratio in basal stem/progenitor cells of parous mice. Separate bioinformatics analyses showed reduced activity for the canonical Wnt transcription factor LEF1/TCF7 and increased activity for the Wnt repressor TCF3. This finding was specific for basal stem/progenitor cells and was associated with downregulation of potentially carcinogenic pathways and a reduction in the proliferation potential of this cell subpopulation in vitro and in vivo. As a possible mechanism for decreased Wnt signaling in basal stem/progenitor cells, we found a more than threefold reduction in the expression of the secreted Wnt ligand Wnt4 in total mammary cells from parous mice, which corresponded to a similar decrease in the proportion of Wnt4-secreting and estrogen/progesterone receptor-positive cells. Because recombinant Wnt4 rescued the proliferation defect of basal stem/progenitor cells in vitro, reduced Wnt4 secretion appears to be causally related to parity-induced alterations of basal stem/progenitor cell properties in mice. CONCLUSIONS: By revealing that parity induces differentiation and downregulates the Wnt/Notch signaling ratio and the in vitro and in vivo proliferation potential of basal stem/progenitor cells in mice, our study sheds light on the long-term consequences of an early pregnancy. Furthermore, it opens the door to future studies assessing whether inhibitors of the Wnt pathway may be used to mimic the parity-induced protective effect against breast cancer.


Assuntos
Diferenciação Celular , Proliferação de Células , Epitélio/patologia , Glândulas Mamárias Animais/citologia , Receptores Notch/metabolismo , Células-Tronco/citologia , Proteínas Wnt/metabolismo , Animais , Antígenos Ly , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Epitélio/metabolismo , Feminino , Citometria de Fluxo , Imunofluorescência , Perfilação da Expressão Gênica , Técnicas Imunoenzimáticas , Glândulas Mamárias Animais/metabolismo , Proteínas de Membrana , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Paridade , Gravidez , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores Notch/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Células-Tronco/metabolismo , Proteínas Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
4.
Breast Cancer Res ; 12(5): R87, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20964822

RESUMO

INTRODUCTION: Normal and neoplastic breast tissues are comprised of heterogeneous populations of epithelial cells involving various degrees of maturation and differentiation. While cultured cell lines have been derived from both normal and malignant tissues, it remains unclear whether they retain a similar cellular heterogeneity as to that found within breast tissues. METHODS: We used 12 reduction mammoplasty tissues, 15 primary breast cancer tissues, and 20 human breast epithelial cell lines (16 cancer lines, 4 normal lines) to perform flow cytometry for CD44, CD24, epithelial cell adhesion molecule (EpCAM), and CD49f expression as well as immunohistochemistry, and in vivo tumor xenograft formation studies to extensively analyze the molecular and cellular characteristics of breast epithelial cell lineages. RESULTS: Human breast tissues contain four distinguishable epithelial differentiation states (two luminal phenotypes and two basal phenotypes) that differ on the basis of CD24, EpCAM and CD49f expression. Primary human breast cancer tissues also contain these four cellular states, but in altered proportions compared to normal tissues. In contrast, cultured cancer cell lines are enriched for rare basal and mesenchymal phenotypes, which are normally present in small numbers within human tissues. Similarly, cultured normal human mammary epithelial cell lines were enriched for rare basal and mesenchymal phenotypes that represent a minor fraction of cells within reduction mammoplasty tissues. Although normal human mammary epithelial cell lines exhibited features of bi-potent progenitor cells they were unable to differentiate into mature luminal breast epithelial cells. CONCLUSIONS: As a group breast cancer cell lines represent the heterogeneity of human breast tumors, but individually they exhibit increased lineage-restricted profiles that fall short of truly representing the intratumoral heterogeneity of individual breast tumors. Additionally, normal human mammary epithelial cell lines fail to retain much of the cellular diversity found in human breast tissues and are enriched for differentiation states that are a minority in breast tissues, although they do exhibit features of bi-potent basal progenitor cells. These findings suggest that collections of cell lines representing multiple cell types can be used to model the cellular heterogeneity of tissues.


Assuntos
Neoplasias da Mama/patologia , Mama/citologia , Células Epiteliais/citologia , Animais , Antígenos de Neoplasias/análise , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/genética , Mama/patologia , Neoplasias da Mama/genética , Antígeno CD24/análise , Moléculas de Adesão Celular/análise , Diferenciação Celular , Linhagem Celular Tumoral , Molécula de Adesão da Célula Epitelial , Células Epiteliais/patologia , Feminino , Expressão Gênica , Humanos , Receptores de Hialuronatos/análise , Integrina alfa6/análise , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
5.
J Cell Physiol ; 217(2): 468-77, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18561256

RESUMO

The three Akt isoforms differ in their ability to transduce oncogenic signals initiated by the Neu and PyMT oncogenes in mammary epithelia. As a result, ablation of Akt1 inhibits and ablation of Akt2 accelerates mammary tumor development by both oncogenes, while ablation of Akt3 is phenotypically almost neutral. Since the risk of breast cancer development in humans correlates with multiple late pregnancies, we embarked on a study to determine whether individual Akt isoforms also differ in their ability to transduce hormonal and growth factor signals during pregnancy, lactation and post-lactation involution. The results showed that the ablation of Akt1 delays the differentiation of the mammary epithelia during pregnancy and lactation, and that the ablation of Akt2 has the opposite effect. Finally, ablation of Akt3 results in minor defects, but its phenotype is closer to that of the wild type mice. Whereas the phenotype of the Akt1 ablation is cell autonomous, that of Akt2 is not. The ablation of Akt1 promotes apoptosis and accelerates involution, whereas the ablation of Akt2 inhibits apoptosis and delays involution. Mammary gland differentiation during pregnancy depends on the phosphorylation of Stat5a, which is induced by prolactin, a hormone that generates signals transduced via Akt. Here we show that the ablation of Akt1, but not the ablation of Akt2 or Akt3 interferes with the phosphorylation of Stat5a during late pregnancy and lactation. We conclude that the three Akt isoforms have different roles in mammary gland differentiation during pregnancy and this may reflect differences in hormonal signaling.


Assuntos
Diferenciação Celular , Células Epiteliais/enzimologia , Lactação , Glândulas Mamárias Animais/enzimologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Apoptose , Proliferação de Células , Sobrevivência Celular , Células Epiteliais/transplante , Feminino , Genótipo , Isoenzimas , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/transplante , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Leite/metabolismo , Fenótipo , Fosforilação , Gravidez , Proteínas Proto-Oncogênicas c-akt/deficiência , Proteínas Proto-Oncogênicas c-akt/genética , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais , Fatores de Tempo
6.
ACS Synth Biol ; 7(2): 474-489, 2018 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-29257672

RESUMO

Cell classifier gene circuits that integrate multiple molecular inputs to restrict the expression of therapeutic outputs to cancer cells have the potential to result in efficacious and safe cancer therapies. Preclinical translation of the hitherto developments requires creating the conditions where the animal model, the delivery platform, in vivo expression levels of the inputs, and the efficacy of the output, all come together to enable detailed evaluation of the fully assembled circuits. Here we show an integrated workflow that addresses these issues and builds the framework for preclinical classifier studies using the design framework of microRNA (miRNA, miR)-based classifier gene circuits. Specifically, we employ HCT-116 colorectal cancer cell xenograft in an experimental mouse metastatic liver tumor model together with Adeno-associated virus (AAV) vector delivery platform. Novel engineered AAV-based constructs are used to validate in vivo the candidate inputs miR-122 and miR-7 and, separately, the cytotoxic output HSV-TK/ganciclovir. We show that while the data are largely consistent with expectations, crucial insights are gained that could not have been obtained in vitro. The results highlight the importance of detailed stepwise interrogation of the experimental parameters as a necessary step toward clinical translation of synthetic gene circuits.


Assuntos
Neoplasias Colorretais , Redes Reguladoras de Genes , Genes Neoplásicos , Neoplasias Hepáticas Experimentais , MicroRNAs , RNA Neoplásico , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Dependovirus , Vetores Genéticos , Humanos , Neoplasias Hepáticas Experimentais/genética , Neoplasias Hepáticas Experimentais/metabolismo , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Neoplásico/genética , RNA Neoplásico/metabolismo
7.
Cytoskeleton (Hoboken) ; 70(10): 635-50, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23804571

RESUMO

Tumor initiation and progression are accompanied by complex changes in the cytoarchitecture that at the cellular level involve remodeling of the cytoskeleton. We report on the impact of a mutant ß-actin (G245D-actin) on cell structure and multicellular assembly properties. To appreciate the effects of the Gly245Asp substitution on the organization of the actin cytoskeleton, we examined the polymerization properties of G245D-actin in vitro by pyrene polymerization assays and total internal reflection fluorescence microscopy (TIRF). The mutant actin on its own has a significantly reduced polymerization efficiency compared to native actin but also modifies the polymerization of actin in copolymerization experiments. Comparison of the structure of Rat-2 fibroblasts and a stably transfected derivate called Rat-2-sm9 revealed the effects of G245D-actin in a cellular environment. The overall actin levels in Rat-2-sm9 show a 1.6-fold increase with similar amounts of mutant and wild-type actin. G245D-actin expression renders Rat-2-sm9 cells highly tumorigenic in nude mice. In Rat-2-sm9 monolayers, G245D-actin triggers the formation of extensive membrane ruffles, which is a characteristic feature of many transformed cells. To approximate complex cell-cell and cell-matrix interactions that occur in tumors and might modulate the effects of G245D-actin, we extended our studies to scaffold-free 3D spheroid cultures. Bright field and scanning electron microscopy (SEM) show that Rat-2-sm9 and Rat-2 cells share essential features of spheroid formation and compaction. However, the resulting spheroids exhibit distinct phenotypes that differ mainly in surface structure and size. The systematic comparison of transformed and normal spheroids by SEM provides new insights into scaffold-free fibroblast spheroid formation.


Assuntos
Actinas/metabolismo , Carcinogênese/patologia , Forma Celular , Fibroblastos/patologia , Mutação/genética , Esferoides Celulares/patologia , Citoesqueleto de Actina/metabolismo , Animais , Carcinogênese/metabolismo , Proliferação de Células , Fibroblastos/metabolismo , Fibroblastos/ultraestrutura , Camundongos , Camundongos Nus , Proteínas Mutantes/metabolismo , Polimerização , Coelhos , Ratos , Esferoides Celulares/metabolismo , Esferoides Celulares/ultraestrutura , Fibras de Estresse/metabolismo
8.
Cancer Cell ; 22(6): 796-811, 2012 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-23238015

RESUMO

Hyperactive PI3K/mTOR signaling is prevalent in human malignancies and its inhibition has potent antitumor consequences. Unfortunately, single-agent targeted cancer therapy is usually short-lived. We have discovered a JAK2/STAT5-evoked positive feedback loop that dampens the efficacy of PI3K/mTOR inhibition. Mechanistically, PI3K/mTOR inhibition increased IRS1-dependent activation of JAK2/STAT5 and secretion of IL-8 in several cell lines and primary breast tumors. Genetic or pharmacological inhibition of JAK2 abrogated this feedback loop and combined PI3K/mTOR and JAK2 inhibition synergistically reduced cancer cell number and tumor growth, decreased tumor seeding and metastasis, and also increased overall survival of the animals. Our results provide a rationale for combined targeting of the PI3K/mTOR and JAK2/STAT5 pathways in triple-negative breast cancer, a particularly aggressive and currently incurable disease.


Assuntos
Neoplasias da Mama/metabolismo , Janus Quinase 2/antagonistas & inibidores , Metástase Neoplásica/tratamento farmacológico , Inibidores de Fosfoinositídeo-3 Quinase , Fator de Transcrição STAT5/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/antagonistas & inibidores , Animais , Neoplasias da Mama/tratamento farmacológico , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Humanos , Proteínas Substratos do Receptor de Insulina/metabolismo , Interleucina-8/metabolismo , Janus Quinase 2/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos SCID , Fosfatidilinositol 3-Quinases/metabolismo , Receptores de Interleucina-8A/metabolismo , Fator de Transcrição STAT5/metabolismo , Serina-Treonina Quinases TOR/metabolismo
9.
Cancer Res ; 72(11): 2705-13, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22467173

RESUMO

Estrogen has a central role in the genesis and progression of breast cancers whether they are positive or negative for the estrogen receptor (ER). While therapies that disrupt estrogen biosynthesis or ER activity can treat these diseases in postmenopausal women, in younger women where ovarian function remains intact, these anti-estrogen therapies are not as effective. Moreover, emerging clinical evidence suggests that estrogen may promote other cancers. Thus, circulating estrogens may participate in cancer pathogenesis in ways that are not yet understood. In this study, we show that estrogen can promote the outgrowth of murine xenograft tumors established from patient-derived ER-negative breast cancer cells by influencing the mobilization and recruitment of a proangiogenic population of bone marrow-derived myeloid cells. ERα expression was necessary and sufficient in the bone marrow-derived cells themselves to promote tumor formation in response to estrogen. Our findings reveal a novel way in which estrogen promotes tumor formation, with implications for the development and application of anti-estrogen therapies to treat cancer in premenopausal women.


Assuntos
Células da Medula Óssea/efeitos dos fármacos , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/patologia , Estradiol/farmacologia , Monócitos/efeitos dos fármacos , Neovascularização Patológica/etiologia , Receptores de Estrogênio/fisiologia , Animais , Células da Medula Óssea/fisiologia , Neoplasias da Mama/química , Movimento Celular/efeitos dos fármacos , Feminino , Humanos , Camundongos , Camundongos Knockout , Monócitos/fisiologia , Receptores de Estrogênio/análise
10.
PLoS One ; 6(9): e24605, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21957456

RESUMO

Fibroblasts are important in orchestrating various functions necessary for maintaining normal tissue homeostasis as well as promoting malignant tumor growth. Significant evidence indicates that fibroblasts are functionally heterogeneous with respect to their ability to promote tumor growth, but markers that can be used to distinguish growth promoting from growth suppressing fibroblasts remain ill-defined. Here we show that human breast fibroblasts are functionally heterogeneous with respect to tumor-promoting activity regardless of whether they were isolated from normal or cancerous breast tissues. Rather than significant differences in fibroblast marker expression, we show that fibroblasts secreting abundant levels of prostaglandin (PGE2), when isolated from either reduction mammoplasty or carcinoma tissues, were both capable of enhancing tumor growth in vivo and could increase the number of cancer stem-like cells. PGE2 further enhanced the tumor promoting properties of fibroblasts by increasing secretion of IL-6, which was necessary, but not sufficient, for expansion of breast cancer stem-like cells. These findings identify a population of fibroblasts which both produce and respond to PGE2, and that are functionally distinct from other fibroblasts. Identifying markers of these cells could allow for the targeted ablation of tumor-promoting and inflammatory fibroblasts in human breast cancers.


Assuntos
Mama/patologia , Dinoprostona/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Fenótipo , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Neoplasias da Mama/cirurgia , Linhagem Celular Tumoral , Separação Celular , Feminino , Humanos , Interleucina-6/metabolismo , Mamoplastia , Camundongos , Transdução de Sinais
11.
Cell Stem Cell ; 8(2): 149-63, 2011 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-21295272

RESUMO

Women with inherited mutations in the BRCA1 gene have increased risk of developing breast cancer but also exhibit a predisposition for the development of aggressive basal-like breast tumors. We report here that breast epithelial cells derived from patients harboring deleterious mutations in BRCA1 (BRCA1(mut /+) give rise to tumors with increased basal differentiation relative to cells from BRCA1+/+ patients. Molecular analysis of disease-free breast tissues from BRCA1(mut /+) patients revealed defects in progenitor cell lineage commitment even before cancer incidence. Moreover, we discovered that the transcriptional repressor Slug is an important functional suppressor of human breast progenitor cell lineage commitment and differentiation and that it is aberrantly expressed in BRCA1(mut /+) tissues. Slug expression is necessary for increased basal-like phenotypes prior to and after neoplastic transformation. These findings demonstrate that the genetic background of patient populations, in addition to affecting incidence rates, significantly impacts progenitor cell fate commitment and, therefore, tumor phenotype.


Assuntos
Neoplasias da Mama/metabolismo , Predisposição Genética para Doença/genética , Células-Tronco/citologia , Adulto , Animais , Neoplasias da Mama/genética , Feminino , Citometria de Fluxo , Genes BRCA1 , Humanos , Imuno-Histoquímica , Camundongos , Camundongos SCID , Pessoa de Meia-Idade , Dados de Sequência Molecular , Fenótipo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição da Família Snail , Células-Tronco/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
12.
Cancer Cell ; 17(1): 65-76, 2010 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-20129248

RESUMO

Transplantation studies have demonstrated the existence of mammary progenitor cells with the ability to self-renew and regenerate a functional mammary gland. Although these progenitors are the likely targets for oncogenic transformation, correlating progenitor populations with certain oncogenic stimuli has been difficult. Cyclin D1 is required for lobuloalveolar development during pregnancy and lactation as well as MMTV-ErbB2- but not MMTV-Wnt1-mediated tumorigenesis. Using a kinase-deficient cyclin D1 mouse, we identified two functional mammary progenitor cell populations, one of which is the target of MMTV-ErbB2. Moreover, cyclin D1 activity is required for the self-renewal and differentiation of mammary progenitors because its abrogation leads to a failure to maintain the mammary epithelial regenerative potential and also results in defects in luminal lineage differentiation.


Assuntos
Diferenciação Celular/fisiologia , Ciclina D1/metabolismo , Glândulas Mamárias Animais/citologia , Neoplasias Mamárias Experimentais/metabolismo , Células-Tronco/citologia , Animais , Antígenos Virais de Tumores/genética , Antígenos Virais de Tumores/metabolismo , Feminino , Citometria de Fluxo , Imunofluorescência , Perfilação da Expressão Gênica , Técnicas de Introdução de Genes , Imuno-Histoquímica , Glândulas Mamárias Animais/metabolismo , Neoplasias Mamárias Experimentais/genética , Camundongos , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Células-Tronco/enzimologia , Proteínas do Core Viral/genética , Proteínas do Core Viral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA