Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 163
Filtrar
1.
Development ; 147(5)2020 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-32098763

RESUMEN

17ß-Estradiol induces the postnatal development of mammary gland and influences breast carcinogenesis by binding to the estrogen receptor ERα. ERα acts as a transcription factor but also elicits rapid signaling through a fraction of ERα expressed at the membrane. Here, we have used the C451A-ERα mouse model mutated for the palmitoylation site to understand how ERα membrane signaling affects mammary gland development. Although the overall structure of physiological mammary gland development is slightly affected, both epithelial fragments and basal cells isolated from C451A-ERα mammary glands failed to grow when engrafted into cleared wild-type fat pads, even in pregnant hosts. Similarly, basal cells purified from hormone-stimulated ovariectomized C451A-ERα mice did not produce normal outgrowths. Ex vivo, C451A-ERα basal cells displayed reduced matrix degradation capacities, suggesting altered migration properties. More importantly, C451A-ERα basal cells recovered in vivo repopulating ability when co-transplanted with wild-type luminal cells and specifically with ERα-positive luminal cells. Transcriptional profiling identified crucial paracrine luminal-to-basal signals. Altogether, our findings uncover an important role for membrane ERα expression in promoting intercellular communications that are essential for mammary gland development.


Asunto(s)
Epitelio/metabolismo , Receptor alfa de Estrógeno/biosíntesis , Glándulas Mamarias Animales/embriología , Comunicación Paracrina/fisiología , Animales , Células Epiteliales/metabolismo , Células Epiteliales/trasplante , Estradiol/metabolismo , Receptor alfa de Estrógeno/genética , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Lipoilación/fisiología , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/trasplante , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal
2.
Breast Cancer Res ; 21(1): 1, 2019 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-30611295

RESUMEN

BACKGROUND: To obtain a deep understanding of the mechanism by which breast cancer develops, the genes involved in tumorigenesis should be analyzed in vivo. Mouse mammary gland can regenerate completely from a mammary stem cell (MaSC), which enables us to analyze the effect of gene expression and repression on tumorigenesis in mammary gland regenerated from genetically manipulated MaSCs. Although lentiviral and retroviral systems have usually been applied for gene transduction into MaSCs, they are associated with difficulty in introducing long, repeated, or transcriptional termination sequences. There is thus a need for an easier and quicker gene delivery system. METHODS: We devised a new system for gene delivery into MaSCs using the piggyBac transposon vectors and electroporation. Compared with viral systems, this system enables easier and quicker transfection of even long, repeated, or transcriptional termination DNA sequences. We designed gene expression vectors of the transposon system, equipped with a luciferase (Luc) expression cassette for monitoring gene transduction into regenerative mammary gland in mice by in-vivo imaging. A doxycycline (Dox)-inducible system was also integrated for expressing the target gene after mammary regeneration to mimic the actual mechanism of tumorigenesis. RESULTS: With this new gene delivery system, genetically manipulated mammary glands were successfully reconstituted even though the vector size was > 200 kb and even in the presence of DNA elements such as promoters and transcription termination sequences, which are major obstacles to viral vector packaging. They differentiated correctly into both basal and luminal cells, and showed normal morphological change and milk production after pregnancy, as well as self-renewal capacity. Using the Tet-On system, gene expression can be controlled by the addition of Dox after mammary reconstitution. In a case study using polyoma-virus middle T antigen (PyMT), oncogene-induced tumorigenesis was achieved. The histological appearance of the tumor was highly similar to that of the mouse mammary tumor virus-PyMT transgenic mouse model. CONCLUSIONS: With this system, gene transduction in the mammary gland can be easily and quickly achieved, and gene expression can be controlled by Dox administration. This system for genetic manipulation could be useful for analyzing genes involved in breast cancer.


Asunto(s)
Diferenciación Celular/genética , Ingeniería Genética/métodos , Glándulas Mamarias Animales/fisiología , Neoplasias Mamarias Experimentales/genética , Células Madre/fisiología , Animales , Línea Celular , Elementos Transponibles de ADN/genética , Proteínas de Unión al ADN/genética , Doxiciclina/administración & dosificación , Femenino , Fibroblastos , Regulación de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/trasplante , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Cultivo Primario de Células/métodos , Transfección/métodos
3.
Development ; 144(7): 1317-1327, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28275010

RESUMEN

Patched 1 (Ptch1) has epithelial, stromal and systemic roles in murine mammary gland organogenesis, yet specific functions remain undefined. Cre-recombinase-mediated Ptch1 ablation in mammary epithelium increased proliferation and branching, but did not phenocopy transgenic expression of activated smoothened (SmoM2). The epithelium showed no evidence of canonical hedgehog signaling, and hyperproliferation was not blocked by smoothened (SMO) inhibition, suggesting a non-canonical function of PTCH1. Consistent with this possibility, nuclear localization of cyclin B1 was increased. In non-epithelial cells, heterozygous Fsp-Cre-mediated Ptch1 ablation increased proliferation and branching, with dysplastic terminal end buds (TEB) and ducts. By contrast, homozygous Ptch1 ablation decreased proliferation and branching, producing stunted ducts filled with luminal cells showing altered ovarian hormone receptor expression. Whole-gland transplantation into wild-type hosts or estrogen/progesterone treatment rescued outgrowth and hormone receptor expression, but not the histological changes. Bone marrow transplantation failed to rescue outgrowth. Ducts of Fsp-Cre;Ptch1fl/fl mice were similar to Fsp-Cre;SmoM2 ducts, but Fsp-Cre;SmoM2 outgrowths were not stunted, suggesting that the histology might be mediated by Smo in the local stroma, with systemic Ptch1 required for ductal outgrowth and proper hormone receptor expression in the mammary epithelium.


Asunto(s)
Epitelio/metabolismo , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/crecimiento & desarrollo , Morfogénesis , Receptor Patched-1/metabolismo , Animales , Trasplante de Médula Ósea , Proliferación Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Epitelio/efectos de los fármacos , Estrógenos/farmacología , Femenino , Proteínas Hedgehog/metabolismo , Integrasas/metabolismo , Glándulas Mamarias Animales/efectos de los fármacos , Glándulas Mamarias Animales/trasplante , Ratones , Modelos Biológicos , Morfogénesis/efectos de los fármacos , Mutación/genética , Células Mieloides/efectos de los fármacos , Células Mieloides/metabolismo , Progesterona/farmacología , Transducción de Señal/efectos de los fármacos , Receptor Smoothened/metabolismo
4.
BMC Cancer ; 15: 562, 2015 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-26228788

RESUMEN

BACKGROUND: The mammary glands of pigs share many functional and morphological similarities with the breasts of humans, raising the potential of their utility for research into the mechanisms underlying normal mammary function and breast carcinogenesis. Here we sought to establish a model for the efficient manipulation and transformation of porcine mammary epithelial cells (pMEC) in vitro and tumor growth in vivo. METHODS: We utilized a vector encoding the red florescent protein tdTomato to transduce populations of pMEC from Yorkshire -Hampshire crossbred female pigs in vitro and in vivo. Populations of primary pMEC were then separated by FACS using markers to distinguish epithelial cells (CD140a-) from stromal cells (CD140a+), with or without further enrichment for basal and luminal progenitor cells (CD49f+). These separated pMEC populations were transduced by lentivirus encoding murine polyomavirus T antigens (Tag) and tdTomato and engrafted to orthotopic or ectopic sites in immunodeficient NOD.Cg-Prkdc (scid) Il2rg (tm1Wjl) /SzJ (NSG) mice. RESULTS: We demonstrated that lentivirus effectively transduces pMEC in vitro and in vivo. We further established that lentivirus can be used for oncogenic-transformation of pMEC ex vivo for generating mammary tumors in vivo. Oncogenic transformation was confirmed in vitro by anchorage-independent growth, increased cell proliferation, and expression of CDKN2A, cyclin A2 and p53 alongside decreased phosphorylation of Rb. Moreover, Tag-transformed CD140a- and CD140a-CD49f + pMECs developed site-specific tumors of differing histopathologies in vivo. CONCLUSIONS: Herein we establish a model for the transduction and oncogenic transformation of pMEC. This is the first report describing a porcine model of mammary epithelial cell tumorigenesis that can be applied to the study of human breast cancers.


Asunto(s)
Antígenos Transformadores de Poliomavirus/genética , Transformación Celular Viral/genética , Lentivirus/genética , Glándulas Mamarias Animales/trasplante , Neoplasias Mamarias Experimentales/patología , Poliomavirus/inmunología , Animales , Proliferación Celular , Células Cultivadas , Femenino , Regulación Neoplásica de la Expresión Génica , Vectores Genéticos , Técnicas In Vitro , Lentivirus/fisiología , Glándulas Mamarias Animales/patología , Glándulas Mamarias Animales/virología , Neoplasias Mamarias Experimentales/etiología , Poliomavirus/genética , Porcinos
5.
Stem Cells Transl Med ; 2(3): 199-203, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23408103

RESUMEN

Mammary gland reconstitution experiments, as well as lineage tracing experiments, have provided evidence for the existence of adult mammary stem cells (MaSCs). In addition, cell sorting techniques for specific cell surface markers (CD24(+)CD29(H)CD49f(H)Sca1(-)) have been used to prospectively isolate MaSC-enriched populations. Although these markers enrich for cell subpopulations that harbor MaSCs, they do not identify regenerative stem cells uniquely. Here, we report that MaSCs can be further defined by the property of cell size. Fluorescence-activated cell sorting was used to analyze sizing beads and further separate populations of cells with varying degrees of forward scatter (FSC). Cells with a low FSC that were approximately <10 µm in size lacked outgrowth potential and failed to reconstitute the mammary gland when transplanted into the cleared fat pads of syngeneic mice. In contrast, cells >10 µm in size with a higher FSC had increased outgrowth potential as compared with lineage-negative (LIN(-)) control cells. Limiting dilution transplantation assays indicated that the repopulating ability of LIN(-)CD24(+)CD29(H) cells that were >10 µm in size was significantly increased as compared with cells marked by CD24 and CD29 alone. These results suggest that MaSCs can be further isolated by sorting based on size/FSC. These findings have critical implications for understanding mammary gland stem cell biology, an important requisite step for understanding the etiology of breast cancer.


Asunto(s)
Separación Celular , Tamaño de la Célula , Células Epiteliales/citología , Glándulas Mamarias Animales/citología , Células Madre/citología , Animales , Biomarcadores/metabolismo , Antígeno CD24/metabolismo , Linaje de la Célula , Proliferación Celular , Separación Celular/métodos , Células Epiteliales/metabolismo , Células Epiteliales/trasplante , Femenino , Citometría de Flujo , Integrina beta1/metabolismo , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/trasplante , Ratones , Ratones Transgénicos , Esferoides Celulares , Trasplante de Células Madre , Células Madre/metabolismo , Factores de Tiempo
6.
Cell Rep ; 3(1): 70-8, 2013 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-23352663

RESUMEN

Mammary epithelial stem cells are vital to tissue expansion and remodeling during various phases of postnatal mammary development. Basal mammary epithelial cells are enriched in Wnt-responsive cells and can reconstitute cleared mammary fat pads upon transplantation into mice. Lgr5 is a Wnt-regulated target gene and was identified as a major stem cell marker in the small intestine, colon, stomach, and hair follicle, as well as in kidney nephrons. Here, we demonstrate the outstanding regenerative potential of a rare population of Lgr5-expressing (Lgr5(+)) mammary epithelial cells (MECs). We found that Lgr5(+) cells reside within the basal population, are superior to other basal cells in regenerating functional mammary glands (MGs), are exceptionally efficient in reconstituting MGs from single cells, and exhibit regenerative capacity in serial transplantations. Loss-of-function and depletion experiments of Lgr5(+) cells from transplanted MECs or from pubertal MGs revealed that these cells are not only sufficient but also necessary for postnatal mammary organogenesis.


Asunto(s)
Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/metabolismo , Organogénesis , Receptores Acoplados a Proteínas G/metabolismo , Animales , Animales Recién Nacidos , Antígeno CD24/metabolismo , Toxina Diftérica/farmacología , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Queratina-14/metabolismo , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/trasplante , Ratones , Ratones Endogámicos C57BL , Regeneración/efectos de los fármacos , Maduración Sexual/efectos de los fármacos , Análisis de la Célula Individual , Tamoxifeno/farmacología
7.
Dev Biol ; 373(1): 95-106, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23103586

RESUMEN

Bone morphogenetic proteins (BMPs) are involved in embryonic mammary gland (MG) development and can be dysregulated in breast cancer. However, the role BMPs play in the postnatal MG remains virtually unknown. BMPs are potent morphogens that are involved in cell fate determination, proliferation, apoptosis and adult tissue homeostasis. Twisted gastrulation (TWSG1) is a secreted BMP binding protein that modulates BMP ligand availability in the extracellular space. Here we investigate the consequences of TWSG1 deletion on development of the postnatal MG. At puberty, Twsg1 is expressed in the myoepithelium and in a subset of body cells of the terminal end buds. In the mature duct, Twsg1 expression is primarily restricted to the myoepithelial layer. Global deletion of Twsg1 leads to a delay in ductal elongation, reduced secondary branching, enlarged terminal end buds, and occluded lumens. This is associated with an increase in luminal epithelial cell number and a decrease in apoptosis. In the MG, pSMAD1/5/8 level and the expression of BMP target genes are reduced, consistent with a decrease in BMP signaling. GATA-3, which is required for luminal identity, is reduced in Twsg1(-/-) MGs, which may explain why K14 positive cells, which are normally restricted to the myoepithelial layer, are found within the luminal compartment and shed into the lumen. In summary, regulation of BMP signaling by TWSG1 is required for normal ductal elongation, branching of the ductal tree, lumen formation, and myoepithelial compartmentalization in the postnatal MG.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Glándulas Mamarias Animales/crecimiento & desarrollo , Proteínas/metabolismo , Transducción de Señal/fisiología , Animales , Apoptosis/fisiología , Western Blotting , Bromodesoxiuridina , Línea Celular , Proliferación Celular , Epitelio/metabolismo , Femenino , Galactósidos , Regulación del Desarrollo de la Expresión Génica/genética , Técnicas de Sustitución del Gen , Hematoxilina , Inmunohistoquímica , Indoles , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/trasplante , Ratones , Ratones Noqueados , Proteínas/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Cancer Cell ; 21(5): 626-641, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22624713

RESUMEN

Basal-like breast cancers (BLBC) express a luminal progenitor gene signature. Notch receptor signaling promotes luminal cell fate specification in the mammary gland, while suppressing stem cell self-renewal. Here we show that deletion of Lfng, a sugar transferase that prevents Notch activation by Jagged ligands, enhances stem/progenitor cell proliferation. Mammary-specific deletion of Lfng induces basal-like and claudin-low tumors with accumulation of Notch intracellular domain fragments, increased expression of proliferation-associated Notch targets, amplification of the Met/Caveolin locus, and elevated Met and Igf-1R signaling. Human BL breast tumors, commonly associated with JAGGED expression, elevated MET signaling, and CAVEOLIN accumulation, express low levels of LFNG. Thus, reduced LFNG expression facilitates JAG/NOTCH luminal progenitor signaling and cooperates with MET/CAVEOLIN basal-type signaling to promote BLBC.


Asunto(s)
Neoplasias de la Mama/enzimología , Caveolinas/metabolismo , Transformación Celular Neoplásica/metabolismo , Glicosiltransferasas/metabolismo , Glándulas Mamarias Animales/enzimología , Neoplasias Mamarias Experimentales/enzimología , Células Madre Neoplásicas/enzimología , Proteínas Proto-Oncogénicas c-met/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteínas de Unión al Calcio/metabolismo , Caveolinas/genética , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Células Cultivadas , Claudinas/metabolismo , Bases de Datos Genéticas , Femenino , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Glicosiltransferasas/deficiencia , Glicosiltransferasas/genética , Humanos , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína Jagged-1 , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/patología , Glándulas Mamarias Animales/trasplante , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Persona de Mediana Edad , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/trasplante , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Proto-Oncogénicas c-met/genética , Receptor IGF Tipo 1/metabolismo , Receptores Notch/metabolismo , Proteínas Serrate-Jagged , Transducción de Señal
9.
Semin Cell Dev Biol ; 23(5): 583-90, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22425744

RESUMEN

While most organs undergo development in utero, the mouse mammary gland orchestrates five major developmental stages following birth: pre-puberty, puberty, pregnancy, lactation, and involution. Induced by both local and systemic factors, these five developmental stages transpire with dramatic alterations in glandular morphology and cellular function. As an experimental system, the mammary gland provides remarkable accessibility to processes regulating stem cell function, hormone response, and epithelial-stromal-extracellular matrix interactions. This review will provide a historical perspective of the unique in vitro and in vivo techniques used to study the mammary gland and how these methods have provided valuable insight into the biology of this organ.


Asunto(s)
Glándulas Mamarias Animales , Animales , Clonación Molecular , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/trasplante , Trasplante de Células Madre , Células Madre/citología , Transgenes
10.
Nature ; 479(7372): 189-93, 2011 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-21983963

RESUMEN

The mammary epithelium is composed of several cell lineages including luminal, alveolar and myoepithelial cells. Transplantation studies have suggested that the mammary epithelium is maintained by the presence of multipotent mammary stem cells. To define the cellular hierarchy of the mammary gland during physiological conditions, we performed genetic lineage-tracing experiments and clonal analysis of the mouse mammary gland during development, adulthood and pregnancy. We found that in postnatal unperturbed mammary gland, both luminal and myoepithelial lineages contain long-lived unipotent stem cells that display extensive renewing capacities, as demonstrated by their ability to clonally expand during morphogenesis and adult life as well as undergo massive expansion during several cycles of pregnancy. The demonstration that the mammary gland contains different types of long-lived stem cells has profound implications for our understanding of mammary gland physiology and will be instrumental in unravelling the cells at the origin of breast cancers.


Asunto(s)
Linaje de la Célula , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/crecimiento & desarrollo , Células Madre/citología , Envejecimiento , Animales , Diferenciación Celular , Trasplante de Células , Epitelio , Femenino , Homeostasis , Lactancia/fisiología , Glándulas Mamarias Animales/fisiología , Glándulas Mamarias Animales/trasplante , Ratones , Células Madre Multipotentes/citología , Embarazo , Células Madre/metabolismo
11.
Dev Biol ; 360(1): 1-10, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21945077

RESUMEN

P190A and p190B Rho GTPase activating proteins (GAPs) are essential genes that have distinct, but overlapping roles in the developing nervous system. Previous studies from our laboratory demonstrated that p190B is required for mammary gland morphogenesis, and we hypothesized that p190A might have a distinct role in the developing mammary gland. To test this hypothesis, we examined mammary gland development in p190A-deficient mice. P190A expression was detected by in situ hybridization in the developing E14.5day embryonic mammary bud and within the ducts, terminal end buds (TEBs), and surrounding stroma of the developing virgin mammary gland. In contrast to previous results with p190B, examination of p190A heterozygous mammary glands demonstrated that p190A deficiency disrupted TEB morphology, but did not significantly delay ductal outgrowth indicating haploinsufficiency for TEB development. To examine the effects of homozygous deletion of p190A, embryonic mammary buds were rescued by transplantation into the cleared fat pads of SCID/Beige mice. Complete loss of p190A function inhibited ductal outgrowth in comparison to wildtype transplants (51% vs. 94% fat pad filled). In addition, the transplantation take rate of p190A deficient whole gland transplants from E18.5 embryos was significantly reduced compared to wildtype transplants (31% vs. 90%, respectively). These results suggest that p190A function in both the epithelium and stroma is required for mammary gland development. Immunostaining for p63 demonstrated that the myoepithelial cell layer is disrupted in the p190A deficient glands, which may result from the defective cell adhesion between the cap and body cell layers detected in the TEBs. The number of estrogen- and progesterone receptor-positive cells, as well as the expression levels of these receptors was increased in p190A deficient outgrowths. These data suggest that p190A is required in both the epithelial and stromal compartments for ductal outgrowth and that it may play a role in mammary epithelial cell differentiation.


Asunto(s)
Proteínas Activadoras de GTPasa/fisiología , Glándulas Mamarias Animales/embriología , Glándulas Mamarias Animales/fisiología , Proteínas Represoras/fisiología , Animales , Secuencia de Bases , Cartilla de ADN/genética , Epitelio/embriología , Femenino , Proteínas Activadoras de GTPasa/deficiencia , Proteínas Activadoras de GTPasa/genética , Regulación del Desarrollo de la Expresión Génica , Heterocigoto , Homocigoto , Hibridación in Situ , Glándulas Mamarias Animales/trasplante , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Embarazo , Receptores de Esteroides/metabolismo , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Células del Estroma/citología
12.
Cancer Cell ; 19(5): 640-51, 2011 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-21575864

RESUMEN

Tissue microenvironment is an important determinant of carcinogenesis. We demonstrate that ionizing radiation, a known carcinogen, affects cancer frequency and characteristics by acting on the microenvironment. Using a mammary chimera model in which an irradiated host is transplanted with oncogenic Trp53 null epithelium, we show accelerated development of aggressive tumors whose molecular signatures were distinct from tumors arising in nonirradiated hosts. Molecular and genetic approaches show that TGFß mediated tumor acceleration. Tumor molecular signatures implicated TGFß, and genetically reducing TGFß abrogated the effect on latency. Surprisingly, tumors from irradiated hosts were predominantly estrogen receptor negative. This effect was TGFß independent and linked to mammary stem cell activity. Thus, the irradiated microenvironment affects latency and clinically relevant features of cancer through distinct and unexpected mechanisms.


Asunto(s)
Neoplasias de la Mama/etiología , Transformación Celular Neoplásica/efectos de la radiación , Células Epiteliales/efectos de la radiación , Glándulas Mamarias Animales/efectos de la radiación , Neoplasias Inducidas por Radiación/etiología , Microambiente Tumoral/efectos de la radiación , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Relación Dosis-Respuesta en la Radiación , Células Epiteliales/metabolismo , Células Epiteliales/patología , Células Epiteliales/trasplante , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Glándulas Mamarias Animales/trasplante , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Neoplasias Inducidas por Radiación/genética , Neoplasias Inducidas por Radiación/metabolismo , Neoplasias Inducidas por Radiación/patología , Quimera por Radiación , Tiempo de Reacción , Receptores de Estrógenos/deficiencia , Factores de Tiempo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Carga Tumoral , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética , Irradiación Corporal Total
13.
Biosci Biotechnol Biochem ; 75(3): 550-5, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21389616

RESUMEN

The interaction between mammary epithelial and stromal tissue is considered to be important in breast tissue development. In this study, we developed a transplantation procedure for the mammary stromal fibroblastic cell line (MSF) to examine its life in vivo. First we established MSF cells which stably expressed lacZ (lacZ/MSF) and had characteristics of mammary stromal cells. The lacZ/MSF cells were then transplanted into a cleared mammary fat pad of syngenic mice with and without mammary primary epithelial organoids. Whole mount X-gal and carmine staining of the transplants revealed that a number of undifferentiated lacZ/MSF cells survived around the mammary epithelial tissue when transplanted with organoids. These results indicate that transplantation of MSF cells into mammary fat pad was accomplished by co-transplantation with primary mammary organoids. Finally, we discuss the application of transplantation procedure for in vivo studies of the mammary stromal tissue development and stromal-epithelial interactions.


Asunto(s)
Fibroblastos/trasplante , Glándulas Mamarias Animales/trasplante , Organoides/trasplante , Comunicación Paracrina , Células del Estroma/trasplante , Tejido Adiposo/citología , Tejido Adiposo/crecimiento & desarrollo , Tejido Adiposo/metabolismo , Animales , Compuestos Azo/análisis , Carmín/análisis , Línea Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Epitelio/crecimiento & desarrollo , Epitelio/metabolismo , Femenino , Fibroblastos/química , Fibroblastos/citología , Fibroblastos/metabolismo , Galactósidos/genética , Galactósidos/metabolismo , Expresión Génica , Inmunohistoquímica , Indoles/metabolismo , Queratinas/análisis , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/crecimiento & desarrollo , Ratones , Organoides/citología , Células del Estroma/química , Células del Estroma/citología , Células del Estroma/metabolismo , Transformación Genética , Trasplante Isogénico
14.
Dev Biol ; 349(2): 125-36, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20974122

RESUMEN

We generated a transgenic (Tg)-mouse model expressing a dominant negative-(DN)-RARα, (RARαG303E) under adipocytes-specific promoter to explore the paracrine role of adipocyte retinoic acid receptors (RARs) in mammary morphogenesis. Transgenic adipocytes had reduced level of RARα, ß and γ, which coincided with a severely underdeveloped pubertal and mature ductal tree with profoundly decreased epithelial cell proliferation. Transplantation experiments of mammary epithelium and of whole mammary glands implicated a fat-pad dependent paracrine mechanism in the stunted phenotype of the epithelial ductal tree. Co-cultures of primary adipocytes, or in vitro differentiated adipocyte cell line, with mammary epithelium showed that when activated, adipocyte-RARs contribute to generation of secreted proliferative and pro-migratory factors. Gene expression microarrays revealed a large number of genes regulated by adipocyte-RARs. Among them, pleiotrophin (PTN) was identified as the paracrine effectors of epithelial cell migration. Its expression was found to be strongly inhibited by DN-RARα, an inhibition relieved by pharmacological doses of all-trans retinoic acid (atRA) in culture and in vivo. Moreover, adipocyte-PTHR, another atRA responsive gene, was found to be an up-stream regulator of PTN. Overall, these results support the existence of a novel paracrine loop controlled by adipocyte-RAR that regulates the mammary ductal tree morphogenesis.


Asunto(s)
Adipocitos/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Glándulas Mamarias Animales/embriología , Morfogénesis/fisiología , Comunicación Paracrina/fisiología , Receptores de Ácido Retinoico/metabolismo , Células 3T3-L1 , Animales , Proteínas Portadoras/metabolismo , Medios de Cultivo Condicionados/química , Citocinas/metabolismo , Cartilla de ADN/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Immunoblotting , Inmunohistoquímica , Glándulas Mamarias Animales/trasplante , Ratones , Ratones Transgénicos , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tretinoina/farmacología
15.
Breast Cancer Res ; 12(5): R79, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20923554

RESUMEN

INTRODUCTION: Retinoic acid signaling pathways are disabled in human breast cancer suggesting a controlling role in normal mammary growth that might be lost in tumorigenesis. We tested a single receptor isotype, RARα1, for its role in mouse mammary gland morphogenesis and MMTV-wnt1-induced oncogenesis. METHODS: The role of RARα1 in mammary morphogenesis was tested in RARα1-knockout (KO) mice and in mammary tumorigenesis in bi-genic (RARα1/KO crossed with MMTV-wnt1) mice. We used whole mounts analysis, stem cells/progenitor quantification, mammary gland repopulation, Q-PCR, test of tumor-free survival, tumor fragments and cell transplantation. RESULTS: In 2 genetic backgrounds (129/Bl-6 and FVB) the neo-natal RARα1/KO-mammary epithelial tree was 2-fold larger and the pubertal tree had 2-fold more branch points and 5-fold more mature end buds, a phenotype that was predominantly epithelial cell autonomous. The stem/progenitor compartment of the RARα1/KO mammary, defined as CD24(low)/ALDH(high activity) was increased by a median 1.7 fold, but the mammary stem cell (MaSC)-containing compartment, (CD24(low)/CD29(high)), was larger (~1.5 fold) in the wt-glands, and the mammary repopulating ability of the wt-gland epithelium was ~2-fold greater. In MMTV-wnt1 transgenic glands the progenitor (CD24(low)/ALDH(high activity)) content was 2.6-fold greater than in the wt and was further increased in the RARα1/KO-wnt1 glands. The tumor-free survival of RARα1/KO-wnt1 mice was significantly (p=0.0002, Kaplan Meier) longer, the in vivo growth of RARα1/KO-wnt1 transplanted tumor fragments was significantly (p=0.01) slower and RARα1/KO-wnt1 tumors cell suspension produced tumors after much longer latency. CONCLUSIONS: In vitamin A-replete mice, RARα1 is required to maintain normal mammary morphogenesis, but paradoxically, also efficient tumorigenesis. While its loss increases the density of the mammary epithelial tree and the content of luminal mammary progenitors, it appears to reduce the size of the MaSC-containing compartment, the mammary repopulating activity, and to delay significantly the MMTV-wnt1-mammary tumorigenesis. Whether the delay in tumorigenesis is solely due to a reduction in wnt1 target cells or due to additional mechanisms remains to be determined. These results reveal the intricate nature of the retinoid signaling pathways in mammary development and carcinogenesis and suggest that a better understanding will be needed before retinoids can join the armament of effective anti- breast cancer therapies.


Asunto(s)
Transformación Celular Neoplásica , Glándulas Mamarias Animales/crecimiento & desarrollo , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/metabolismo , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Proteína Wnt1/metabolismo , Familia de Aldehído Deshidrogenasa 1 , Animales , Antígeno CD24/análisis , Femenino , Integrina beta1/análisis , Isoenzimas/metabolismo , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/trasplante , Virus del Tumor Mamario del Ratón/genética , Virus del Tumor Mamario del Ratón/fisiología , Ratones , Ratones Noqueados , Morfogénesis , Retinal-Deshidrogenasa/metabolismo , Receptor alfa de Ácido Retinoico , Transducción de Señal , Células Madre/citología , Proteína Wnt1/genética
16.
J Vis Exp ; (40)2010 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-20548284

RESUMEN

This article describes and compares the fat pad clearance procedure developed by DeOme KB et al. and the sparing procedure developed by Brill B et al., followed by the mammary epithelial transplant procedure. The mammary transplant procedure is widely used by mammary biologists because it takes advantage of the fact that significant development of the mammary epithelium doesn't occur until after puberty. At 3 weeks of age, growth of the mammary epithelial tree is confined to the vicinity of the nipple and the fat pad is largely devoid of mammary epithelium, but by 7 weeks of age the epithelial ductal tree extends throughout the entire fat pad. Therefore, if this small portion of the fat pad containing epithelium, the region between the nipple and the lymph node, is removed at 3 weeks of age, the endogenous epithelium will never populate the mammary fat pad and the fat pad is described as "cleared". At this time, mammary epithelium from another source can be transplanted in the cleared fat pad where it has the potential to extend mammary ductal trees through out the fat pad. This procedure has been utilized in many experimental models including the examination of tumor phenotype in transgenic mammary epithelial tissue without the confounding effects of genotype on the entire animal, in the identification of mammary stem cells by transplanting cells in limited dilution, determining if hyperplastic nodules proceed to mammary tumors, and to assess the effect of prior hormone exposure on the behavior of the mammary epithelium. Three week old host mice are anesthetized, cleaned and restrained on a surgical stage. A mid-sagittal incision is made through the skin, but not the peritoneum, extending from the pubis to the sternum. Oblique cuts are made through the skin from the mid-sagittal incision across the pelvis toward each leg. The skin is pulled away from the peritoneum to expose the 4th inguinal mammary gland. The fat pad is cleared by removing the fat pad tissue anterior to the lymph node. Epithelium fragments or epithelial cells are transplanted into the remaining cleared fat pad and the mouse is closed.


Asunto(s)
Glándulas Mamarias Animales/trasplante , Trasplante de Tejidos/métodos , Animales , Epitelio/trasplante , Femenino , Ratones
17.
PLoS One ; 5(5): e10691, 2010 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-20502530

RESUMEN

BACKGROUND: Considerable evidence indicates that heparan sulfate is essential for the development of tissues consisting of branching ducts and tubules. However, there are few examples where specific sulfate residues regulate a specific stage in the formation of such tissues. METHODOLOGY/PRINCIPAL FINDINGS: We examined the role of heparan sulfation in mammary gland branching morphogenesis, lactation and lobuloalveolar development by inactivation of heparan sulfate GlcNAc N-deacetylase/N-sulfotransferase genes (Ndst) in mammary epithelial cells using the Cre-loxP system. Ndst1 deficiency resulted in an overall reduction in glucosamine N-sulfation and decreased binding of FGF to mammary epithelial cells in vitro and in vivo. Mammary epithelia lacking Ndst1 underwent branching morphogenesis, filling the gland with ductal tissue by sexual maturity to the same extent as wildtype epithelia. However, lobuloalveolar expansion did not occur in Ndst1-deficient animals, resulting in insufficient milk production to nurture newly born pups. Lactational differentiation of isolated mammary epithelial cells occurred appropriately via stat5 activation, further supporting the notion that the lack of milk production was due to lack of expansion of the lobuloalveoli. CONCLUSIONS/SIGNIFICANCE: These findings demonstrate a selective, highly penetrant, cell autonomous effect of Ndst1-mediated sulfation on lobuloalveolar development.


Asunto(s)
Células Epiteliales/enzimología , Células Epiteliales/patología , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/patología , Sulfotransferasas/deficiencia , Animales , Femenino , Silenciador del Gen , Marcación de Gen , Integrasas/metabolismo , Lactancia , Glándulas Mamarias Animales/trasplante , Virus del Tumor Mamario del Ratón/metabolismo , Ratones , Morfogénesis , Coloración y Etiquetado , Sulfotransferasas/metabolismo , Azufre/metabolismo
18.
EMBO J ; 29(5): 956-68, 2010 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-20075866

RESUMEN

PI 3-kinase enhancer A (PIKE-A) is critical for the activation of Akt signalling, and has an essential function in promoting cancer cell survival. However, its physiological functions are poorly understood. Here, we show that PIKE-A directly associates with both signal transducer and activator of transcription 5a (STAT5a) and prolactin (PRL) receptor, which is essential for PRL-provoked STAT5a activation and the subsequent gene transcription. Depletion of PIKE-A in HC11 epithelial cells diminished PRL-induced STAT5 activation and cyclin D1 expression, resulting in profoundly impaired cell proliferation in vitro. To confirm the function of PIKE-A in PRL signalling in vivo, we generated PIKE knockout (PIKE-/-) mice. PIKE-/- mice displayed a severe lactation defect that was characterized by enhanced apoptosis and impaired proliferation of mammary epithelial cells. At parturition, STAT5 activation and cyclin D1 expression were substantially reduced in the mammary epithelium of PIKE-/- mice. The defective mammary gland development in PIKE-/- mice was rescued by overexpression of a mammary-specific cyclin D1 transgene. These data establish a critical function for PIKE-A in mediating PRL functions.


Asunto(s)
GTP Fosfohidrolasas/metabolismo , Glándulas Mamarias Animales/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Prolactina/farmacología , Factor de Transcripción STAT5/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Western Blotting , Línea Celular , Proliferación Celular , Ciclina D1/metabolismo , Células Epiteliales/citología , Células Epiteliales/fisiología , Femenino , GTP Fosfohidrolasas/genética , Expresión Génica , Genotipo , Inmunoprecipitación , Etiquetado Corte-Fin in Situ , Lactancia/genética , Lactancia/fisiología , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/trasplante , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Embarazo , Unión Proteica/efectos de los fármacos , Receptores de Prolactina/metabolismo
19.
J Pathol ; 220(1): 34-44, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19927317

RESUMEN

The contribution of cancer cell-intrinsic and -extrinsic factors to metastatic breast cancer is still poorly understood, hampering development of novel therapeutic strategies that decrease breast cancer mortality. Cre/loxP-based conditional mouse models of breast cancer present unique opportunities to study sporadic tumour formation and progression in a controlled setting. Unfortunately, the generation of mouse strains carrying multiple mutant alleles needed for such studies is very time-consuming. Moreover, conditional mouse tumour models do not permit independent manipulation of tumour cell-intrinsic and -extrinsic factors. Although the latter can be achieved by cleared fat-pad transplantation of mouse mammary epithelial cells (MMECs) from tumour suppressor gene (TSG) knockouts into wild-type or mutant recipients, this procedure is not possible for mutations that cause embryonic lethality or preclude mammary gland development. Here we show that cleared fat-pad transplantations with MMECs isolated from K14cre;Cdh1(F/F); Trp53(F/F) mice expressing Cre recombinase under control of the cytokeratin-14 promoter and carrying conditional null alleles for p53 and E-cadherin (Cdh1) first resulted in the formation of phenotypically normal mammary glands, followed by the development of invasive metastatic mammary tumours. Tumour formation in the recipients mimicked tumour latency, spectrum, morphology, immunophenotype, and metastatic characteristics of the original mammary tumour model. This transplantation system, which can be expanded to other conditional TSG knockouts, permits independent genetic analysis of stromal factors and testing of additional cancer cell-intrinsic mutations that would otherwise be embryonic lethal or require intensive breeding.


Asunto(s)
Transformación Celular Neoplásica/patología , Glándulas Mamarias Animales/trasplante , Neoplasias Mamarias Experimentales/patología , Tejido Adiposo/trasplante , Animales , Cadherinas/metabolismo , Transformación Celular Neoplásica/metabolismo , Células Epiteliales/trasplante , Femenino , Queratina-8/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Noqueados , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas de Neoplasias/metabolismo , Procesos Estocásticos , Vimentina/metabolismo
20.
Genes Dev ; 23(22): 2563-77, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19933147

RESUMEN

The epithelium of the mammary gland exists in a highly dynamic state, undergoing dramatic morphogenetic changes during puberty, pregnancy, lactation, and regression. The recent identification of stem and progenitor populations in mouse and human mammary tissue has provided evidence that the mammary epithelium is organized in a hierarchical manner. Characterization of these normal epithelial subtypes is an important step toward understanding which cells are predisposed to oncogenesis. This review summarizes progress in the field toward defining constituent cells and key molecular regulators of the mammary epithelial hierarchy. Potential relationships between normal epithelial populations and breast tumor subtypes are discussed, with implications for understanding the cellular etiology underpinning breast tumor heterogeneity.


Asunto(s)
Neoplasias de la Mama/patología , Glándulas Mamarias Animales/patología , Glándulas Mamarias Humanas/patología , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/fisiopatología , Diferenciación Celular , Femenino , Regulación de la Expresión Génica , Humanos , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/trasplante , Glándulas Mamarias Humanas/citología , Mutación/genética , Embarazo , Células Madre/citología , Ubiquitina-Proteína Ligasas/genética , Proteínas Wnt/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA