Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Cell Sci ; 130(12): 2018-2025, 2017 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-28455412

RESUMO

Amphiregulin (AREG)-/- mice demonstrate impaired mammary development and form only rudimentary ductal epithelial trees; however, AREG-/- glands are still capable of undergoing alveologenesis and lactogenesis during pregnancy. Transplantation of AREG-/- mammary epithelial cells into cleared mouse mammary fat pads results in a diminished capacity for epithelial growth (∼15%) as compared to that of wild-type mammary epithelial cells. To determine whether estrogen receptor α (ERα, also known as ESR1) and/or AREG signaling were necessary for non-mammary cell redirection, we inoculated either ERα-/- or AREG-/- mammary cells with non-mammary progenitor cells (WAP-Cre/Rosa26LacZ+ male testicular cells or GFP-positive embryonic neuronal stem cells). ERα-/- cells possessed a limited ability to grow or reprogram non-mammary cells in transplanted mammary fat pads. AREG-/- mammary cells were capable of redirecting both types of non-mammary cell populations to mammary phenotypes in regenerating mammary outgrowths. Transplantation of fragments from AREG-reprogrammed chimeric outgrowths resulted in secondary outgrowths in six out of ten fat pads, demonstrating the self-renewing capacity of the redirected non-mammary cells to contribute new progeny to chimeric outgrowths. Nestin was detected at the leading edges of developing alveoli, suggesting that its expression may be essential for lobular expansion.


Assuntos
Anfirregulina/genética , Linhagem da Célula , Reprogramação Celular , Células Epiteliais/citologia , Transdução de Sinais , Animais , Diferenciação Celular , Proliferação de Células , Transplante de Células , Córtex Cerebral/embriologia , Células-Tronco Embrionárias/citologia , Receptor alfa de Estrogênio/genética , Estrogênios/metabolismo , Feminino , Proteínas de Fluorescência Verde/metabolismo , Masculino , Glândulas Mamárias Animais/citologia , Camundongos , Camundongos Nus , Camundongos Transgênicos , Células-Tronco Neurais/citologia , Gravidez , Espermatozoides/metabolismo , Testículo/metabolismo
2.
J Cell Sci ; 127(Pt 1): 27-32, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24190884

RESUMO

We have previously shown that non-mammary and tumorigenic cells can respond to the signals of the mammary niche and alter their cell fate to that of mammary epithelial progenitor cells. Here we tested the hypothesis that paracrine signals from mammary epithelial cells expressing progesterone receptor (PR) are dispensable for redirection of testicular cells, and that re-directed wild-type testicular-derived mammary cells can rescue lobulogenesis of PR-null mammary epithelium by paracrine signaling during pregnancy. We injected PR-null epithelial cells mixed with testicular cells from wild-type adult male mice into cleared fat-pads of recipient mice. The testicular cells were redirected in vivo to mammary epithelial cell fate during regeneration of the mammary epithelium, and persisted in second-generation outgrowths. In the process, the redirected testicular cells rescued the developmentally deficient PR-null cells, signaling them through the paracrine factor RANKL to produce alveolar secretory structures during pregnancy. This is the first demonstration that paracrine signaling required for alveolar development is not required for cellular reprogramming in the mammary gland, and that reprogrammed testicular cells can provide paracrine signals to the surrounding mammary epithelium.


Assuntos
Reprogramação Celular/genética , Células Epiteliais/citologia , Glândulas Mamárias Animais/citologia , Comunicação Parácrina/genética , Receptores de Progesterona/genética , Túbulos Seminíferos/citologia , Tecido Adiposo , Animais , Diferenciação Celular , Células Epiteliais/metabolismo , Células Epiteliais/transplante , Feminino , Expressão Gênica , Injeções , Masculino , Glândulas Mamárias Animais/metabolismo , Camundongos , Gravidez , Progesterona/metabolismo , Ligante RANK/genética , Ligante RANK/metabolismo , Receptores de Progesterona/deficiência , Túbulos Seminíferos/metabolismo , Túbulos Seminíferos/transplante , Transdução de Sinais
3.
J Mammary Gland Biol Neoplasia ; 20(1-2): 93-101, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26362796

RESUMO

Mammotropic hormones and growth factors play a very important role in mammary growth and differentiation. Here, hormones including Estrogen, Progesterone, Prolactin, their cognate receptors, and the growth factor Amphiregulin, are tested with respect to their roles in signaling non-mammary cells from the mouse to redirect to mammary epithelial cell fate(s). This was done in the context of glandular regeneration in pubertal athymic female mice. Our previous studies demonstrated that mammary stem cell niches are recapitulated during gland regeneration in vivo. During this process, cells of exogenous origin cooperate with mammary epithelial cells to form mammary stem cell niches and thus respond to normal developmental signals. In all cases tested with the possible exception of estrogen receptor alpha (ER-α), hormone signaling is dispensable for non-mammary cells to undertake mammary epithelial cell fate(s), proliferate, and contribute progeny to chimeric mammary outgrowths. Importantly, redirected non-mammary cell progeny, regardless of their source, have the ability to self-renew and contribute offspring to secondary mammary outgrowths derived from transplanted chimeric mammary fragments; thus suggesting that some of these cells are capable of mammary stem cell/progenitor functions.


Assuntos
Diferenciação Celular , Células Epiteliais/metabolismo , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo , Anfirregulina/metabolismo , Animais , Proliferação de Células , Estrogênios/metabolismo , Camundongos , Progesterona/metabolismo , Prolactina/metabolismo , Receptores de Progesterona/metabolismo , Células-Tronco/fisiologia
4.
Dev Dyn ; 240(3): 674-81, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21337465

RESUMO

Prominin-1 (Prom1) is recognized as a stem cell marker in several tissues, including blood, neuroepithelium, and gut, and in human and mouse embryos and many cancers. Although Prom1 is routinely used as a marker for isolating stem cells, its biological function remains unclear. Here we use a knockout model to investigate the role of Prom1 in the mammary gland. We demonstrate that complete loss of Prom1 does not affect the regenerative capacity of the mammary epithelium. Surprisingly, we also show that in the absence of Prom1, mammary glands have reduced ductal branching, and an increased ratio of luminal to basal cells. The effects of Prom1 loss in the mammary gland are associated with decreased expression of prolactin receptor and matrix metalloproteinase-3. These experiments reveal a novel, functional role for Prom1 that is not related to stem cell activity, and demonstrate the importance of tissue-specific characterization of putative stem cell markers.


Assuntos
Antígenos CD/metabolismo , Glicoproteínas/metabolismo , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/fisiologia , Morfogênese/fisiologia , Peptídeos/metabolismo , Regeneração/fisiologia , Células-Tronco/metabolismo , Antígeno AC133 , Animais , Antígenos CD/genética , Feminino , Citometria de Fluxo , Glicoproteínas/genética , Glândulas Mamárias Animais/citologia , Camundongos , Camundongos Mutantes , Camundongos Nus , Morfogênese/genética , Peptídeos/genética , Regeneração/genética
5.
Exp Cell Res ; 316(3): 422-32, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19913532

RESUMO

Amphiregulin (AREG), a ligand for epidermal growth factor receptor, is required for mammary gland ductal morphogenesis and mediates estrogen actions in vivo, emerging as an essential growth factor during mammary gland growth and differentiation. The COMMA-D beta-geo (CDbetageo) mouse mammary cell line displays characteristics of normal mammary progenitor cells including the ability to regenerate a mammary gland when transplanted into the cleared fat pad of a juvenile mouse, nuclear label retention, and the capacity to form anchorage-independent mammospheres. We demonstrate that AREG is essential for formation of floating mammospheres by CDbetageo cells and that the mitogen activated protein kinase signaling pathway is involved in AREG-mediated mammosphere formation. Addition of exogenous AREG promotes mammosphere formation in cells where AREG expression is knocked down by siRNA and mammosphere formation by AREG(-/-) mammary epithelial cells. AREG knockdown inhibits mammosphere formation by duct-limited mammary progenitor cells but not lobule-limited mammary progenitor cells. These data demonstrate AREG mediates the function of a subset of mammary progenitor cells in vitro.


Assuntos
Células Epiteliais/citologia , Células Epiteliais/metabolismo , Glicoproteínas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Glândulas Mamárias Animais/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Anfirregulina , Animais , Linhagem Celular Transformada , Proliferação de Células , Família de Proteínas EGF , Células Epiteliais/enzimologia , Feminino , Glicoproteínas/antagonistas & inibidores , Glicoproteínas/deficiência , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , RNA Interferente Pequeno/metabolismo , Transfecção
6.
Proc Natl Acad Sci U S A ; 105(39): 14891-6, 2008 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-18809919

RESUMO

A fundamental issue in stem cell biology is whether adult somatic stem cells are capable of accessing alternate tissue sites and continue functioning as stem cells in the new microenvironment. To address this issue relative to neurogenic stem cells in the mouse mammary gland microenvironment, we mixed wild-type mammary epithelial cells (MECs) with bona fide neural stem cells (NSCs) isolated from WAP-Cre/Rosa26R mice and inoculated them into cleared fat pads of immunocompromised females. Hosts were bred 6-8 weeks later and examined postinvolution. This allowed for mammary tissue growth, transient activation of the WAP-Cre gene, recombination, and constitutive expression of LacZ. The NSCs and their progeny contributed to mammary epithelial growth during ductal morphogenesis, and the Rosa26-LacZ reporter gene was activated by WAP-Cre expression during pregnancy. Some NSC-derived LacZ(+) cells expressed mammary-specific functions, including milk protein synthesis, whereas others adopted myoepithelial cell fates. Thus, NSCs and their progeny enter mammary epithelium-specific niches and adopt the function of similarly endowed mammary cells. This result supports the conclusion that tissue-specific signals emanating from the stroma and from the differentiated somatic cells of the mouse mammary gland can redirect the NSCs to produce cellular progeny committed to MEC fates.


Assuntos
Diferenciação Celular , Glândulas Mamárias Animais/crescimento & desenvolvimento , Células-Tronco Multipotentes/citologia , Neurônios/citologia , Animais , Ciclo Celular , Diferenciação Celular/genética , Células Epiteliais/citologia , Feminino , Genes Reporter , Glândulas Mamárias Animais/citologia , Camundongos , Camundongos Transgênicos , Proteínas do Leite/genética , Morfogênese , Gravidez , Proteínas/genética , RNA não Traduzido , Transplante de Células-Tronco , beta-Galactosidase/genética
7.
Breast Cancer Res ; 12(5): R86, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20964820

RESUMO

INTRODUCTION: During selective segregation of DNA, a cell asymmetrically divides and retains its template DNA. Asymmetric division yields daughter cells whose genome reflects that of the parents', simultaneously protecting the parental cell from genetic errors that may occur during DNA replication. We hypothesized that long-lived epithelial cells are present in immortal, premalignant cell populations, undergo asymmetric division, retain their template DNA strands, and cycle both during allometric growth and during pregnancy. METHODS: The glands of 3-week old immune competent Balb/C female mice were utilized intact or cleared of host epithelium and implanted with ductal-limited, lobule-limited, or alveolar-ductal progenitor cells derived from COMMA-D1 pre-malignant epithelial cells. 5-bromo-2-deoxyuridine (5-BrdU) was administered to identify those cells which retain their template DNA. Nulliparous mice were then either injected with [(3)H]-thymidine ((3)H-TdR) to distinguish 5-BrdU-label retaining cells that enter the cell cycle and euthanized, or mated, injected with (3)H-TdR, and euthanized at various days post-coitus. Sections were stained for estrogen receptor-α(ER-α) or progesterone receptor (PR) via immunohistochemistry. Cells labelled with both 5-BrdU and (3)H-TdR were indicative of label-retaining epithelial cells (LREC). RESULTS: Cells that retained a 5-BrdU label and cells labelled with [(3)H]-thymidine were found in all mice and were typically detected along the branching epithelium of mature mouse mammary glands. Cells containing double-labelled nuclei (LREC) were found in the intact mammary gland of both pregnant and nulliparous mice, and in mammary glands implanted with pre-malignant cells. Double-labelled cells ((3)H-TdR/5-BrdU) represent a small portion of cells in the mammary gland that cycle and retain their template DNA (5-BrdU). Some label-retaining cells were also ER-α or PR positive. LRECs distributed their second label ((3)H-TdR) to daughter cells; and this effect persisted during pregnancy. LRECs, and small focal hyperplasia, were found in all immortalized premalignant mammary implant groups. CONCLUSIONS: The results indicate that a subpopulation of long-lived, label-retaining epithelial cells (LRECs) is present in immortal premalignant cell populations. These LRECs persist during pregnancy, retain their original DNA, and a small percentage express ER-α and PR. We speculate that LRECs in premalignant hyperplasia represent the long-lived (memory) cells that maintain these populations indefinitely.


Assuntos
Divisão Celular Assimétrica/genética , Replicação do DNA , DNA/biossíntese , Células Epiteliais/metabolismo , Glândulas Mamárias Animais/citologia , Animais , Autorradiografia , Bromodesoxiuridina , Células Epiteliais/citologia , Receptor alfa de Estrogênio/análise , Feminino , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Lesões Pré-Cancerosas , Gravidez , Receptores de Progesterona/análise , Células-Tronco/citologia , Células-Tronco/metabolismo , Moldes Genéticos , Timidina , Trítio
8.
Oncotarget ; 11(2): 161-174, 2020 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-32010429

RESUMO

One major foundation of cancer etiology is the process of clonal expansion. The mechanisms underlying the complex process of a single cell leading to a clonal dominant tumor, are poorly understood. Our study aims to analyze mitochondrial DNA (mtDNA) for somatic single nucleotide polymorphisms (SNPs) variants, to determine if they are conserved throughout clonal expansion in mammary tissues and tumors. To test this hypothesis, we took advantage of a mouse mammary tumor virus (MMTV)-infected mouse model (CzechII). CzechII mouse mtDNA was extracted, from snap-frozen normal, hyperplastic, and tumor mammary epithelial outgrowth fragments. Next generation deep sequencing was used to determine if mtDNA "de novo" SNP variants are conserved during serial transplantation of both normal and neoplastic mammary clones. Our results support the conclusion that mtDNA "de novo" SNP variants are selected for and maintained during serial passaging of clonal phenotypically heterogeneous normal cellular populations; neoplastic cellular populations; metastatic clonal cellular populations and in individual tumor transplants, grown from the original metastatic tumor. In one case, a mammary tumor arising from a single cell, within a clonal hyperplastic outgrowth, contained only mtDNA copies, harboring a deleterious "de novo" SNP variant, suggesting that only one mtDNA template may act as a template for all mtDNA copies regardless of cell phenotype. This process has been attributed to "heteroplasmic-shifting". A process that is thought to result from selective pressure and may be responsible for pathogenic mutated mtDNA copies becoming homogeneous in clonal dominant oncogenic tissues.

9.
Oncotarget ; 11(30): 2919-2929, 2020 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-32774772

RESUMO

The influence of breast cancer cells on normal cells of the microenvironment, such as fibroblasts and macrophages, has been heavily studied but the influence of normal epithelial cells on breast cancer cells has not. Here using in vivo and in vitro models we demonstrate the impact epithelial cells and the mammary microenvironment can exert on breast cancer cells. Under specific conditions, signals that originate in epithelial cells can induce phenotypic and genotypic changes in cancer cells. We have termed this phenomenon "cancer cell redirection." Once breast cancer cells are redirected, either in vivo or in vitro, they lose their tumor forming capacity and undergo a genetic expression profile shift away from one that supports a cancer profile towards one that supports a non-tumorigenic epithelial profile. These findings indicate that epithelial cells and the normal microenvironment influence breast cancer cells and that under certain circumstances restrict proliferation of tumorigenic cells.

10.
Mech Dev ; 159: 103565, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31336167

RESUMO

Long-label retention has been used by many to prove Cairns' immortal strand hypothesis and to identify potential stem cells. Here, we describe two strategies using 5-ethynl-2'-deoxyuridine (EdU) to identify and understand the distribution of long-label-retaining mammary epithelial cells during formation of the mouse mammary ductal system. First, EdU was given upon two consecutive days per week during weeks 4 through 10 and analyzed for label retention at 13 weeks of age. Alternatively, EdU was given for 14 consecutive days beginning at 28 days of age and ending at 42 days of age. Analyses were conducted at >91 days of age (13 weeks). Many more LREC were detected following the second labeling method and their distribution among the subsequently developed ducts. This finding indicated that the early-labeled cells that retained their label were distributed into portions of the gland that developed after the ending of EdU treatment (i.e. 42->91 days). These observations may have important meaning with respect to the previously demonstrated retention of regenerative capacity throughout the mouse mammary gland despite age or reproductive history. These results suggest LREC may represent long-lived progenitor cells that are responsible for mammary gland homeostasis. Additionally, these cells may act as multipotent stem cells capable of mammary gland regeneration upon random fragment transplantation into epithelium-denuded mammary fat pads.


Assuntos
Células Epiteliais/citologia , Células Epiteliais/metabolismo , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Organogênese , Animais , Feminino , Fase G2 , Camundongos Endogâmicos BALB C , Camundongos Nus , Coloração e Rotulagem
11.
Oncotarget ; 10(22): 2118-2135, 2019 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-31040905

RESUMO

Microarray technologies were used to analyze transcriptomes from Comma-Dß and clonal derivatives, SP3 (Lobule-competent) and NSP2 (Lobule-incompetent), during different mouse mammary growth phases: in-vitro, in-vivo 5-weeks, and in-vivo 12-weeks. A differentially expressed gene (DEG) algorithm was used to enrich for genes associated with cellular proliferation, differentiation, cell cycle regulation, and carcinogenesis. A pairwise comparison analysis, of SP3 vs. NSP2 in-vitro, revealed a total of 45 DEGs significantly up-regulated in SP3. Of the 45 DEGs, only Ccnd1 (Cyclin D1), Id2 (Inhibitor of DNA binding 2) and Sox9 (SRY Box 9) were identified to be associated with cellular proliferation, regulation of G1/S mitotic cell cycle, mammary gland and alveolar development in SP3. During the regenerative growth phase, in-vivo 5-weeks, we identified a total of 545 DEGs. 308 DEGs, of the 545 DEGs, were significantly up-regulated and 237 DEGs were significantly down-regulated in SP3 vs. NSP2. In addition, we identified 9 DEGs significantly up-regulated, within SP3's cell cycle pathway and a persistent overexpression of Cyclin D1, Id2, and Sox9, consistent with our in-vitro study. During the maintenance phase, in-vivo 12-weeks, we identified 407 DEGs. Of these, 336 DEGs were up-regulated, and 71 were down-regulated in SP3 vs. NSP2. Our data shows 15 DEGs significantly up-regulated, simultaneously, affecting 8 signal transducing carcinogenic pathways. In conclusion, increased expression of Cyclin D1, Id2 and Sox9 appear to be important for lobular genesis in SP3. Also, in-vivo 12 week displays increase expression of genes and pathways, involved in tumorigenesis.

12.
Breast Cancer Res ; 10(5): R90, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18950502

RESUMO

INTRODUCTION: During pregnancy the mammary epithelial compartment undergoes extreme proliferation and differentiation, facilitated by stem/progenitor cells. Mouse mammary epithelium in nonpregnant mice contains long label-retaining epithelial cells (LREC) that divide asymmetrically and retain their template DNA strands. The role of LREC during alveogenesis has not been determined. METHODS: We performed immunohistochemistry and autoradiography on murine mammary glands that had been labeled with 5-bromodeoxyuridine (5BrdU) during allometric ductal growth to investigate the co-expression of DNA label retention and estrogen receptor-alpha or progesterone receptor during pregnancy. A second DNA label ([3H]-thymidine) was administered during pregnancy to identify label-retaining cells (LRC), which subsequently enter the cell cycle. Use of this methodology allowed us to investigate the co-localization of 5BrdU with smooth muscle actin, CD31, cytokeratin, and desmin in periductal or peri-acinar LRC in mammary tissue from pregnant mice subsequent to a long chase period in order to identify LRC. RESULTS: Estrogen receptor-alpha positive and progesterone receptor positive cells represented approximately 30% to 40% of the LREC, which is under 1.0% of the epithelial subpopulation. Pregnancy altered the percentage of LREC expressing estrogen receptor-alpha. LRC situated in periductal or peri-acinar positions throughout the gland do not express epithelial, endothelial, or myoepithelial markers, and these undefined LRCs persist throughout pregnancy. Additionally, new cycling LREC ([3H]-thymidine retaining) appear during alveologenesis, and LRC found in other tissue types (for example, endothelium and nerve) within the mammary fat pad become double labeled during pregnancy, which indicates that they may also divide asymmetrically. CONCLUSIONS: Our findings support the premise that there is a subpopulation of LREC in the mouse mammary gland that persists during alveologenesis. These cells react to hormonal cues during pregnancy and enter the cell cycle while continuing to retain, selectively, their original template DNA. In addition, nonepithelial LRC are found in periductal or peri-acinar positions. These LRC also enter the cell cycle during pregnancy. During alveologenesis, newly created label-retaining ([3H]-thymidine) epithelial cells appear within the expanding alveoli and continue to cycle and retain their original template DNA ([3H]-thymidine) strands, as determined by a second pulse of 5BrdU.


Assuntos
Células-Tronco Adultas/citologia , Anáfase , Cromátides/química , DNA/genética , Glândulas Mamárias Animais/metabolismo , Prenhez/metabolismo , Actinas/análise , Células-Tronco Adultas/metabolismo , Animais , Biomarcadores , Bromodesoxiuridina/análise , Bromodesoxiuridina/farmacocinética , Linhagem da Célula , DNA/análise , Desmina/análise , Receptor alfa de Estrogênio/análise , Feminino , Queratinas/análise , Glândulas Mamárias Animais/citologia , Camundongos , Modelos Genéticos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/análise , Gravidez , Receptores de Progesterona/análise , Moldes Genéticos , Trítio/análise , Trítio/farmacocinética
14.
Breast Cancer Res ; 9(4): R42, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17626637

RESUMO

INTRODUCTION: Int6 has been shown to be an interactive participant with the protein translation initiation complex eIF3, the COP9 signalosome and the regulatory lid of the 26S proteasome. Insertion of mouse mammary tumor virus into the Int6 locus creates a C-terminally truncated form of the protein. Expression of the truncated form of Int6 (Int6sh) in stably transfected human and mouse mammary epithelial cell lines leads to cellular transformation. In addition, decreased expression of Int6/eIF3e is observed in approximately one third of all human breast carcinomas. METHODS: To validate that Int6sh has transforming activity in vivo, a transgenic mouse model was designed using the whey acidic protein (Wap) promoter to target expression of truncated Int6 to differentiating alveolar epithelial cells in the mammary gland. Microarray analyses were performed on normal, premalignant and malignant WapInt6sh expressing tissues. RESULTS: Mammary tumors developed in 42% of WapInt6sh heterozygous parous females at an average age of 18 months. In WapInt6sh mice, the contralateral mammary glands from both tumorous and non-tumorous tissues contained widespread focal alveolar hyperplasia. Only 4% of WapInt6sh non-breeding females developed tumors by 2 years of age. The Wap promoter is active only during estrus in the mammary tissue of cycling non-pregnant mice. Microarray analyses of mammary tissues demonstrated that Int6sh expression in the alveolar tissue altered the mammary transcriptome in a specific manner that was detectable even in the first pregnancy. This Int6sh-specific transcriptome pattern subsequently persisted in both the Int6sh-expressing alveolar hyperplasia and mammary tumors. These observations are consistent with the conclusion that WapInt6sh-expressing alveolar cells survive involution following the cessation of lactation, and subsequently give rise to the mammary tumors that arise in aging multiparous females. CONCLUSION: These observations provide direct in vivo evidence that mammary-specific expression of the Int6sh truncation leads to persistence of alveolar hyperplasia with the accompanying increased predisposition to mammary tumorigenesis.


Assuntos
Transformação Celular Neoplásica , Epitélio/metabolismo , Fator de Iniciação 3 em Eucariotos/metabolismo , Hiperplasia/etiologia , Glândulas Mamárias Animais/metabolismo , Neoplasias Mamárias Experimentais/etiologia , Animais , Biomarcadores Tumorais/metabolismo , Fator de Iniciação 3 em Eucariotos/genética , Feminino , Perfilação da Expressão Gênica , Humanos , Hiperplasia/patologia , Masculino , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Transgênicos , Proteínas do Leite/genética , Análise de Sequência com Séries de Oligonucleotídeos , Plasmídeos/genética , Gravidez , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Deleção de Sequência , Transfecção , Células Tumorais Cultivadas
15.
Methods Mol Biol ; 1501: 277-289, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27796959

RESUMO

This chapter considers the techniques necessary and required for the reprogramming of exogenous stem/progenitor cell populations towards a mammary epithelial cell fate. The protocols describe how to isolate cells from alternate mouse organs such as testicles of male mice and mix them with mammary cells to generate chimeric glands comprised of male and female epithelial cells that are fully competent. During the reformation of mammary stem cell niches by dispersed epithelial cells, in the context of the intact epithelium-free mammary stroma, non-mammary cells are sequestered and reprogrammed to perform mammary epithelial cell functions including those ascribed to mammary stem/progenitor cells. This therefore is a powerful technique for the redirection of cells from other organs/cancer cells to a normal mammary phenotype.


Assuntos
Células Epiteliais/fisiologia , Epitélio/fisiologia , Glândulas Mamárias Animais/fisiologia , Células-Tronco/fisiologia , Animais , Diferenciação Celular/fisiologia , Feminino , Masculino , Camundongos , Camundongos Transgênicos/fisiologia , Nicho de Células-Tronco/fisiologia
16.
Sci Rep ; 7: 40196, 2017 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-28071703

RESUMO

Previously, we demonstrated the ability of the normal mammary microenvironment (niche) to direct non-mammary cells including testicular and embryonic stem cells (ESCs) to adopt a mammary epithelial cell (MEC) fate. These studies relied upon the interaction of transplanted normal MECs with non-mammary cells within the mammary fat-pads of recipient mice that had their endogenous epithelium removed. Here, we tested whether acellular mammary extracellular matrix (mECM) preparations are sufficient to direct differentiation of testicular-derived cells and ESCs to form functional mammary epithelial trees in vivo. We found that mECMs isolated from adult mice and rats were sufficient to redirect testicular derived cells to produce normal mammary epithelial trees within epithelial divested mouse mammary fat-pads. Conversely, ECMs isolated from omental fat and lung did not redirect testicular cells to a MEC fate, indicating the necessity of tissue specific components of the mECM. mECM preparations also completely inhibited teratoma formation from ESC inoculations. Further, a phenotypically normal ductal outgrowth resulted from a single inoculation of ESCs and mECM. To the best of our knowledge, this is the first demonstration of a tissue specific ECM driving differentiation of cells to form a functional tissue in vivo.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/fisiologia , Matriz Extracelular/metabolismo , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Animais , Camundongos , Ratos
17.
Oncogene ; 24(4): 552-60, 2005 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-15580303

RESUMO

A parity-induced mammary population, marked by beta-galactosidase expression conditionally activated through cre-lox recombinase originates in WAP-Cre/Rosa-lox-STOP-lox-LacZ (WAP-Cre/Rosa-LacZ) female mice during pregnancy, lactation and involution. During subsequent pregnancies, these parity-induced mammary epithelial cells (PI-MEC) proliferated to produce new secretory acini composed of secretory luminal cells and myoepithelium. In serial transplantation assays, PI-MEC were able to self-renew over several transplant generations and to contribute significantly to the resulting mammary outgrowths. In limiting dilution transplantation, they proliferated to produce both luminal and myoepithelial cells, comprised both lobule-limited and duct-limited epithelial outgrowths, and differentiated into all the cellular subtypes recognized in murine mammary epithelium. TGF-beta1 expression from the whey acidic protein promoter (WAP) in triply transgenic females did not prevent the appearance of PI-MEC after pregnancy despite the absence of full lactation or their ability to proliferate and produce progeny with diverse cellular fates in situ upon subsequent pregnancies. However, in transplants from triple transgenic parous females, the WAP-TGF-beta1-positive PI-MEC did not contribute to the newly recapitulated mammary outgrowths, suggesting that they were incapable of expansive cellular proliferation (self-renewal). This result is consistent with our earlier publication that WAP-TGF-beta1 expression in mammary epithelium induces premature stem cell senescence in mammary transplants and decreases mammary cancer risk in mouse mammary tumor virus (MMTV)-infected females even after multiple pregnancies.


Assuntos
Células Epiteliais/citologia , Glândulas Mamárias Animais/citologia , Paridade/fisiologia , Células-Tronco Pluripotentes/citologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Linhagem da Célula , Proliferação de Células , Transplante de Células , Células Cultivadas , Células Epiteliais/metabolismo , Feminino , Integrases/metabolismo , Óperon Lac/genética , Masculino , Glândulas Mamárias Animais/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas do Leite/genética , Proteínas do Leite/metabolismo , Células-Tronco Pluripotentes/metabolismo , Fator de Crescimento Transformador beta1 , Proteínas Virais/metabolismo
18.
Aging (Albany NY) ; 8(7): 1353-63, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27347776

RESUMO

It has been proposed that the erosion of telomere length is a limiting factor in replicative capacity and important in cell senescence. To determine if this activity was essential in the mouse mammary gland in vivo, we serially transplanted mammary fragments from wild type (TER+/+), heterozygous (TER+/-), and homozygous (TER-/-) mammary tissues into the cleared mammary fat pads of immune-compromised nude mice. Individual implants from both homozygous and heterozygous TER null outgrowths showed growth senescence beginning at transplant generation two, earlier than implants from TER+/+ mammary glands which continued to show growth. This result suggests that either mammary epithelial stem cells maintain their telomere length in order to self renew, or that the absence or reduction of telomerase template results in more frequent death/extinction of stem cells during symmetric divisions. A third possibility is the inability of signaling cells in the niche to replicate resulting in reduction of the maintenance signals necessary for stem cell renewal. Consistent with this, examination of senescent outgrowths revealed the absence of estrogen receptor alpha (ERα+) epithelium although progesterone receptor (PR+) cells were abundant. Despite their inability to establish mammary growth in vivo, TER+/- cells were able to direct neural stem cells to mammary cell fates.


Assuntos
Reprogramação Celular/fisiologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Células-Tronco Neurais/metabolismo , Telomerase/metabolismo , Animais , Receptor alfa de Estrogênio/metabolismo , Feminino , Glândulas Mamárias Animais/metabolismo , Camundongos , Camundongos Knockout , Camundongos Nus , Receptores de Progesterona/metabolismo , Telomerase/genética
19.
Cell Prolif ; 36 Suppl 1: 3-15, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14521512

RESUMO

An entire mammary epithelial outgrowth, capable of full secretory differentiation, may be comprised of the progeny of a single cellular antecedent. This conclusion is based upon the maintenance of retroviral insertion sites within the somatic DNA of successive transplant generations derived from a single mammary fragment. In addition, dissociation of these clonal dominant glands and implantation of dispersed cells at limiting dilution demonstrated that both duct-limited and lobule-limited outgrowths were developed, as well as complete, fully differentiated glands. Thus, transplantation has revealed three distinct mammary epithelial progenitors in the mouse. Similar studies have extended this observation to rat mammary tissue. Recently, using cre-lox conditional activation of reporter genes, a new epithelial progenitor, specific for mammary secretory epithelium in postlactation females has been uncovered. In situ, these cells were shown to regenerate secretory lobules upon successive pregnancies. In transplant studies, they demonstrated the capacity for self-renewal and contributed to the new generation of all of the known epithelial cell types among mammary epithelium. In limiting dilution, the parity-induced progenitors were capable of engendering lobule-limited and duct-limited outgrowths in their entirety, but not completely developed glands. Serial transplant studies indicate that these progenitors have a significant but limited capacity for self-renewal.


Assuntos
Células Epiteliais/transplante , Glândulas Mamárias Animais/citologia , Transplante de Células-Tronco , Animais , Células Epiteliais/citologia , Células-Tronco/citologia
20.
Mech Ageing Dev ; 123(11): 1505-19, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12425957

RESUMO

The proliferative lifespan of mammary stem cells was examined in serially transplanted clonal-dominant epithelial populations. Five successive transplant generations were done. The epithelial cell number in each outgrowth expands approximately 500-fold in nulliparous hosts and approximately 10000-fold in impregnated hosts. Despite this, all resulting mammary outgrowths showed lineal identity with the original. Growth senescence was observed in some implants beginning at the third generation in impregnated recipients. The ability of an individual implant to support ductal morphogenesis and also secretory lobule development decayed at independent rates. Individual implants from a single clonal-dominant outgrowth occasionally gave rise to markedly different ductal development within the same host indicating an epithelial cell autonomous mechanism in ductal patterning. Both premalignant and malignant populations appeared focally within the aging transplants. These populations were also lineally related to the original outgrowth supporting the conclusion that the primary growth was derived clonally from one or a few lineally related antecedents. The premalignant and malignant descendant populations no longer exhibit growth senescence suggesting that they are supported by a perpetually self-renewing progenitor. Our evidence indicates that a single mammary cell may have the capacity to self-renew through five transplant generations. Even some sixth generation implants show vigorous growth.


Assuntos
Glândulas Mamárias Animais/patologia , Vírus do Tumor Mamário do Camundongo , Células-Tronco/patologia , Animais , Contagem de Células , Diferenciação Celular , Divisão Celular , Linhagem Celular , Senescência Celular/fisiologia , Células Clonais , Células Epiteliais/patologia , Células Epiteliais/transplante , Células Epiteliais/virologia , Feminino , Hiperplasia , Glândulas Mamárias Animais/virologia , Neoplasias Mamárias Experimentais/patologia , Camundongos , Fenótipo , Células-Tronco Pluripotentes/patologia , Lesões Pré-Cancerosas/patologia , Gravidez , Infecções por Retroviridae/patologia , Células-Tronco/fisiologia , Células-Tronco/virologia , Células Tumorais Cultivadas , Infecções Tumorais por Vírus/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA