Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Breast Cancer Res ; 24(1): 30, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35440032

RESUMEN

BACKGROUND: Parathyroid hormone-related protein (PTHrP) is required for embryonic breast development and has important functions during lactation, when it is produced by alveolar epithelial cells and secreted into the maternal circulation to mobilize skeletal calcium used for milk production. PTHrP is also produced by breast cancers, and GWAS studies suggest that it influences breast cancer risk. However, the exact functions of PTHrP in breast cancer biology remain unsettled. METHODS: We developed a tetracycline-regulated, MMTV (mouse mammary tumor virus)-driven model of PTHrP overexpression in mammary epithelial cells (Tet-PTHrP mice) and bred these mice with the MMTV-PyMT (polyoma middle tumor-antigen) breast cancer model to analyze the impact of PTHrP overexpression on normal mammary gland biology and in breast cancer progression. RESULTS: Overexpression of PTHrP in luminal epithelial cells caused alveolar hyperplasia and secretory differentiation of the mammary epithelium with milk production. This was accompanied by activation of Stat5 and increased expression of E74-like factor-5 (Elf5) as well as a delay in post-lactation involution. In MMTV-PyMT mice, overexpression of PTHrP (Tet-PTHrP;PyMT mice) shortened tumor latency and accelerated tumor growth, ultimately reducing overall survival. Tumors overproducing PTHrP also displayed increased expression of nuclear pSTAT5 and Elf5, increased expression of markers of secretory differentiation and milk constituents, and histologically resembled secretory carcinomas of the breast. Overexpression of PTHrP within cells isolated from tumors, but not PTHrP exogenously added to cell culture media, led to activation of STAT5 and milk protein gene expression. In addition, neither ablating the Type 1 PTH/PTHrP receptor (PTH1R) in epithelial cells nor treating Tet-PTHrP;PyMT mice with an anti-PTH1R antibody prevented secretory differentiation or altered tumor latency. These data suggest that PTHrP acts in a cell-autonomous, intracrine manner. Finally, expression of PTHrP in human breast cancers is associated with expression of genes involved in milk production and STAT5 signaling. CONCLUSIONS: Our study suggests that PTHrP promotes pathways leading to secretory differentiation and proliferation in both normal mammary epithelial cells and in breast tumor cells.


Asunto(s)
Neoplasias de la Mama , Neoplasias Mamarias Animales , Proteína Relacionada con la Hormona Paratiroidea , Factor de Transcripción STAT5 , Animales , Neoplasias de la Mama/patología , Femenino , Humanos , Lactancia/genética , Glándulas Mamarias Animales , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/metabolismo , Ratones , Proteína Relacionada con la Hormona Paratiroidea/genética , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/metabolismo
2.
Development ; 139(22): 4239-49, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23034629

RESUMEN

Parathyroid hormone-related protein (PTHrP) regulates cell fate and specifies the mammary mesenchyme during embryonic development. Loss of PTHrP or its receptor (Pthr1) abolishes the expression of mammary mesenchyme markers and allows mammary bud cells to revert to an epidermal fate. By contrast, overexpression of PTHrP in basal keratinocytes induces inappropriate differentiation of the ventral epidermis into nipple-like skin and is accompanied by ectopic expression of Lef1, ß-catenin and other markers of the mammary mesenchyme. In this study, we document that PTHrP modulates Wnt/ß-catenin signaling in the mammary mesenchyme using a Wnt signaling reporter, TOPGAL-C. Reporter expression is completely abolished by loss of PTHrP signaling and ectopic reporter activity is induced by overexpression of PTHrP. We also demonstrate that loss of Lef1, a key component of the Wnt pathway, attenuates the PTHrP-induced abnormal differentiation of the ventral skin. To characterize further the contribution of canonical Wnt signaling to embryonic mammary development, we deleted ß-catenin specifically in the mammary mesenchyme. Loss of mesenchymal ß-catenin abolished expression of the TOPGAL-C reporter and resulted in mammary buds with reduced expression of mammary mesenchyme markers and impaired sexual dimorphism. It also prevented the ectopic, ventral expression of mammary mesenchyme markers caused by overexpression of PTHrP in basal keratinocytes. Therefore, we conclude that a mesenchymal, canonical Wnt pathway mediates the PTHrP-dependent specification of the mammary mesenchyme.


Asunto(s)
Glándulas Mamarias Animales/embriología , Mesodermo/embriología , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Vía de Señalización Wnt , Animales , Diferenciación Celular , Femenino , Regulación del Desarrollo de la Expresión Génica , Queratinocitos/metabolismo , Factor de Unión 1 al Potenciador Linfoide/biosíntesis , Glándulas Mamarias Animales/metabolismo , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Ratones Noqueados , Proteína Relacionada con la Hormona Paratiroidea/deficiencia , Proteína Relacionada con la Hormona Paratiroidea/genética , Receptores de Hormona Paratiroidea/biosíntesis , Receptores de Hormona Paratiroidea/deficiencia , Receptores de Hormona Paratiroidea/genética , Trombospondinas/metabolismo , Proteínas Wnt/biosíntesis , Proteínas Wnt/metabolismo , beta Catenina/biosíntesis , beta Catenina/metabolismo
3.
Breast Cancer Res ; 16(6): 487, 2014 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-25467960

RESUMEN

INTRODUCTION: Studies have identified multi-potent stem cells in the adult mammary gland. More recent studies have suggested that the embryonic mammary gland may also contain stem/progenitor cells that contribute to initial ductal development. We were interested in determining whether embryonic cells might also directly contribute to long-lived stem cells that support homeostasis and development in the adult mammary gland. METHODS: We used DNA-label retention to detect long label-retaining cells in the mammary gland. Mouse embryos were labeled with 5-ethynl-2'-deoxyuridine (EdU) between embryonic day 14.5 and embryonic day 18.5 and were subsequently sacrificed and examined for EdU retention at various intervals after birth. EdU retaining cells were co-stained for various lineage markers and identified after fluorescence activated cell sorting analysis of specific epithelial subsets. EdU-labeled mice were subjected to subsequent 5-bromo-2'-deoxyuridine administration to determine whether EdU-labeled cells could re-enter the cell cycle. Finally, EdU-labeled cells were grown under non-adherent conditions to assess their ability to form mammospheres. RESULTS: We demonstrate embryonically-derived, long label-retaining cells (eLLRCs) in the adult mammary gland. eLLRCs stain for basal markers and are enriched within the mammary stem cell population identified by cell sorting. eLLRCs are restricted to the primary ducts near the nipple region. Interestingly, long label retaining cells (labeled during puberty) are found just in front of the eLLRCs, near where the ends of the ducts had been at the time of DNA labeling in early puberty. A subset of eLLRCs becomes mitotically active during periods of mammary growth and in response to ovarian hormones. Finally, we show that eLLRCs are contained within primary and secondary mammospheres. CONCLUSIONS: Our findings suggest that a subset of proliferating embryonic cells subsequently becomes quiescent and contributes to the pool of long-lived mammary stem cells in the adult. eLLRCs can re-enter the cell cycle, produce both mammary lineages and self-renew. Thus, our studies have identified a putative stem/progenitor cell population of embryonic origin. Further study of these cells will contribute to an understanding of how quiescent stem cells are generated during development and how fetal exposures may alter future breast cancer risk in adults.


Asunto(s)
Células Madre Adultas/citología , Células Madre Embrionarias/citología , Glándulas Mamarias Animales/citología , Células Madre Multipotentes/citología , Células Madre Adultas/metabolismo , Animales , Células Madre Embrionarias/metabolismo , Femenino , Glándulas Mamarias Animales/metabolismo , Ratones , Células Madre Multipotentes/metabolismo
4.
J Mammary Gland Biol Neoplasia ; 18(2): 155-63, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23660702

RESUMEN

The first member of the Wnt-family ligands was identified 30 years ago as a factor in mouse mammary tumours whose expression was deregulated due to the promoter activity emanating from the proximal integration of the Mouse Mammary Tumour Virus genome (Nusse and Varmus, Embo J 31:2670-84, 2012). The Wnt-ligands invoke a number of molecular-genetic signalling cascades fundamental to the patterning of developing tissues and organs during embryogenesis as well as during postnatal development. The Wnt-signalling cascade that controls the activities of ß-catenin and the T-cell Factor (Tcf)/Lympoid enhancer factor (Lef1) plays a fundamental role in control of all stages of embryonic mammary gland development. We provide here a brief overview of the known aspects of Wnt-signalling activities in the embryonic mammary gland and its interactions with other signalling cascades in this developing tissue.


Asunto(s)
Glándulas Mamarias Animales/embriología , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Humanas/embriología , Glándulas Mamarias Humanas/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/fisiología , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Proteínas Wnt/genética
5.
Breast Cancer Res ; 14(2): 307, 2012 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-22546075

RESUMEN

Parathyroid hormone-related protein (PTHrP) causes hypercalcemia in cancer patients. PTHrP is required for normal breast development and has been shown to promote bone metastases from breast cancers. However, whether the protein also contributes to the formation of primary tumors has been unclear. Two recent papers suggest it may. First, a report in Nature Genetics identified the PTHrP locus as a new breast cancer susceptibility gene. Second, a paper in Journal of Clinical Investigation demonstrated that PTHrP promotes tumor growth and metastases in MMTV-PyMT mice. These studies implicate PTHrP in the development and growth of primary breast tumors and underscore the need for further research.


Asunto(s)
Neoplasias de la Mama/genética , Cromosomas Humanos Par 12/genética , Cromosomas Humanos Par 21/genética , Sitios Genéticos/genética , Predisposición Genética a la Enfermedad/genética , Neoplasias Mamarias Experimentales/patología , Metástasis de la Neoplasia/genética , Proteína Relacionada con la Hormona Paratiroidea/fisiología , Animales , Femenino , Humanos
6.
Int J Dev Biol ; 50(7): 601-10, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16892173

RESUMEN

Lymphoid Enhancer Factor-1 (Lef1) facilitates the assembly of transcriptional regulatory complexes and mediates nuclear responses to Wnt signals. We determined previously that the mesenchymally restricted, paired-like homeodomain protein Aristaless-like 4 (Alx4) interacts with Lef1 and together alters promoter activity of candidate genes. In order to define their overlapping functions, mice deficient for both Lef1 and Alx4 activity (Lef1-/-/Alx4lstD/lstD) were produced. Whereas embryos lacking either Lef1 or Alx4 activity remain viable up to or after birth, early embryonic lethality results when both factors were absent. No viable Lef1-/-/Alx4lstD/lstD embryos were recovered beyond 9.5 dpc. Between E8.5 and E10, viable Lef1-/-/Alx4lstD/lstD embryos were developmentally delayed 0.5 days relative to littermates of all other genotypes. Principle among the alterations seen in Lef1-/-/Alx4lstD/lstD animals was defective vasculature in both embryonic and extra-embryonic tissues. In the yolk sac, while the vascular network is present, it were greatly diminished and large vitelline vessels were largely absent. Platelet/endothelial cell adhesion molecule (PECAM) staining revealed that the major vessels in the head of compound mutant embryos were absent, while the other vessels were finer than those seen in normal littermates. Pools of blood and pericardial effusion were also apparent in Lef1-/-/Alx4lstD/lstD animals, further indicative of a defective vasculature. These data confirm genetically the interaction between Lef1 and Alx4 and further reveal unknown, overlapping roles for these transcription factors in embryonic vasculogenesis.


Asunto(s)
Desarrollo Embrionario , Proteínas de Homeodominio/metabolismo , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Animales , Cruzamientos Genéticos , Femenino , Técnica del Anticuerpo Fluorescente Directa , Regulación del Desarrollo de la Expresión Génica , Heterocigoto , Proteínas de Homeodominio/genética , Inmunohistoquímica , Factor de Unión 1 al Potenciador Linfoide/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica/embriología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Embarazo , Saco Vitelino/irrigación sanguínea
7.
PLoS One ; 9(5): e90418, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24785493

RESUMEN

Parathyroid hormone-related protein (PTHrP) can be secreted from cells and interact with its receptor, the Type 1 PTH/PTHrP Receptor (PTHR1) in an autocrine, paracrine or endocrine fashion. PTHrP can also remain inside cells and be transported into the nucleus, where its functions are unclear, although recent experiments suggest that it may broadly regulate cell survival and senescence. Disruption of either the PTHrP or PTHR1 gene results in many abnormalities including a failure of embryonic mammary gland development in mice and in humans. In order to examine the potential functions of nuclear PTHrP in the breast, we examined mammary gland development in PTHrP (1-84) knock-in mice, which express a mutant form of PTHrP that lacks the C-terminus and nuclear localization signals and which can be secreted but cannot enter the nucleus. Interestingly, we found that PTHrP (1-84) knock-in mice had defects in mammary mesenchyme differentiation and mammary duct outgrowth that were nearly identical to those previously described in PTHrP-/- and PTHR1-/- mice. However, the mammary buds in PTHrP (1-84) knock-in mice had severe reductions in mutant PTHrP mRNA levels, suggesting that the developmental defects were due to insufficient production of PTHrP by mammary epithelial cells and not loss of PTHrP nuclear function. Examination of the effects of nuclear PTHrP in the mammary gland in vivo will require the development of alternative animal models.


Asunto(s)
Eliminación de Gen , Glándulas Mamarias Animales/embriología , Glándulas Mamarias Animales/metabolismo , Señales de Localización Nuclear/genética , Proteína Relacionada con la Hormona Paratiroidea/biosíntesis , Proteína Relacionada con la Hormona Paratiroidea/genética , Dominios y Motivos de Interacción de Proteínas , Animales , Biomarcadores/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Glándulas Mamarias Animales/crecimiento & desarrollo , Ratones , Ratones Transgénicos , Proteína Relacionada con la Hormona Paratiroidea/química , Fragmentos de Péptidos/genética , Caracteres Sexuales
8.
PLoS One ; 6(11): e27278, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22087279

RESUMEN

PTHrP is necessary for the formation of the embryonic mammary gland and, in its absence, the embryonic mammary bud fails to form the neonatal duct system. In addition, PTHrP is produced by the breast during lactation and contributes to the regulation of maternal calcium homeostasis during milk production. In this study, we examined the role of PTHrP during post-natal mammary development. Using a PTHrP-lacZ transgenic mouse, we surveyed the expression of PTHrP in the developing post-natal mouse mammary gland. We found that PTHrP expression is restricted to the basal cells of the gland during pubertal development and becomes expressed in milk secreting alveolar cells during pregnancy and lactation. Based on the previous findings that overexpression of PTHrP in cap and myoepithelial cells inhibited ductal elongation during puberty, we predicted that ablation of native PTHrP expression in the post-natal gland would result in accelerated ductal development. To address this hypothesis, we generated two conditional models of PTHrP-deficiency specifically targeted to the postnatal mammary gland. We used the MMTV-Cre transgene to ablate the floxed PTHrP gene in both luminal and myoepithelial cells and a tetracycline-regulated K14-tTA;tetO-Cre transgene to target PTHrP expression in just myoepithelial and cap cells. In both models of PTHrP ablation, we found that mammary development proceeds normally despite the absence of PTHrP. We conclude that PTHrP signaling is not required for normal ductal or alveolar development.


Asunto(s)
Glándulas Mamarias Animales/crecimiento & desarrollo , Proteína Relacionada con la Hormona Paratiroidea/fisiología , Animales , Femenino , Expresión Génica , Lactancia , Ratones , Ratones Transgénicos , Proteína Relacionada con la Hormona Paratiroidea/análisis , Proteína Relacionada con la Hormona Paratiroidea/deficiencia , Embarazo , Distribución Tisular
9.
Organogenesis ; 4(2): 116-22, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19279723

RESUMEN

Wnt signals play a critical role in regulating the normal development of the mammary gland and dysregulation of Wnt signaling causes breast cancer. This pathway is involved in the earliest development of the mammary gland in embryos and its role extends through the functional differentiation of the gland during pregnancy. In this review, we summarize the molecular mechanisms through which Wnts regulate mammary gland development in the mouse.

10.
Dev Biol ; 295(1): 219-31, 2006 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16678815

RESUMEN

Inductive reciprocal signaling between mesenchymal and adjacent epithelia gives rise to skin appendages such as hair follicles and mammary glands. Lef1-mediated canonical Wnt signaling is required for morphogenesis of these skin appendages during embryogenesis. In order to define the role of canonical Wnt signaling during early embryonic mammary gland development, we determined the temporal and spatial changes in Wnt signaling during embryogenesis in wild-type and Lef1-deficient embryos harboring a Tcf/Lef1-betagal reporter (TOPGAL) transgene. In contrast to previous studies using TOPGAL mice from a distinct founder, we observe that Wnt signaling acts initially on mesenchymal cells associated with the sequential appearance of mammary placodes. As placode development progresses between 12.5 and 15.5 dpc, Wnt signaling progressively accumulates in the mammary epithelial compartment. By 18.5 dpc, betagal activity is confined to mesenchymal and epithelial cells near the nipple region. In Lef1-deficient embryos, the transition of Wnt signaling from mesenchyme to the mammary epithelia is blocked for placodes #1, 4 and 5 despite the expression of Tcf1 in epithelial cells. These placodes ultimately disappear by 15.5 dpc, while placodes 2 and 3 typically did not form in the absence of Lef1. Progressive loss of placodes 1, 4, and 5 is accompanied by increased apoptosis in mesenchymal cells adjacent to the mammary epithelial placodes. While factors important for embryonic mammary gland development, such as FGF7, are expressed normally in Lef1-deficient animals, one mediator of the Hedgehog (Hh)-signaling pathway is aberrantly expressed. Specifically, Shh, Ihh, and Gli2 are expressed in mammary epithelial cells at levels in Lef1-deficient animals similar to wild-type littermates. However, the signal for Ptc-1 is strongly reduced in mesenchymal cells surrounding the mammary placode in Lef1 mutants relative to wild-type embryos. The loss of Ptc-1, both a receptor for and transcriptional target of Hh signaling, suggests that Hh signaling is blocked in Lef1-deficient embryos. Thus, these data reveal distinct requirements of different mammary placodes for Lef1-dependent Wnt signaling. They further define dynamic changes in which cells integrate Lef1-dependent Wnt signaling during progression of embryonic mammary gland development.


Asunto(s)
Factor de Unión 1 al Potenciador Linfoide/metabolismo , Glándulas Mamarias Animales/embriología , Mesodermo/metabolismo , Proteínas Wnt/metabolismo , Animales , Células Epiteliales/metabolismo , Femenino , Factor 7 de Crecimiento de Fibroblastos/genética , Factor 7 de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog , Factor de Unión 1 al Potenciador Linfoide/genética , Glándulas Mamarias Animales/metabolismo , Mesodermo/citología , Ratones , Ratones Transgénicos , Receptores Patched , Receptor Patched-1 , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Proteínas Wnt/genética
11.
Dev Biol ; 264(1): 153-65, 2003 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-14623238

RESUMEN

The embryonic mammary gland and hair follicle are both derived from the ventral ectoderm, and their development depends on a number of common fundamental developmental pathways. While the Hedgehog (Hh) signaling pathway is required for hair follicle morphogenesis, the role of this pathway during embryonic mammary gland development remains undetermined. We demonstrate here that, unlike the hair follicle, both Shh and Ihh are expressed in the developing embryonic mouse mammary rudiment as early as E12.5. In Shh(-/-) embryos, hair follicle development becomes arrested at an early stage, while the mammary rudiment, which continues to express Ihh, develops in a manner indistinguishable from that of wild-type littermates. The five pairs of mammary buds in Shh(-/-) female embryos exhibit normal branching morphogenesis at E16.5, forming a rudimentary ductal structure identical to wild-type embryonic mammary glands. We further demonstrate that loss of Hh signaling causes altered cyclin D1 expression in the embryonic dermal mesenchyme. Specifically, cyclin D1 is expressed at E14.5 principally in the condensed mesenchymal cells of the presumptive hair follicles and in both mesenchymal and epithelial cells of the mammary rudiments in wild-type and Shh-deficient embryos. By E18.5, robust cyclin D1 expression is maintained in mammary rudiments of both wild-type and Shh-deficient embryos. In hair follicles of wild-type embryos by E18.5, cyclin D1 expression switches to follicular epithelial cells. In contrast, strong cyclin D1 expression is observed principally in the mesenchymal cells of arrested hair follicles in Shh(-/-) embryos at E18.5. These data reveal that, despite the common embryonic origin of hair follicles and mammary glands, distinct patterns of Hh-family expression occur in these two tissues. Furthermore, these data suggest that cyclin D1 expression in the embryonic hair follicle is mediated by both Hh-independent and Hh-dependent mechanisms.


Asunto(s)
Folículo Piloso/embriología , Glándulas Mamarias Animales/embriología , Transactivadores/metabolismo , Animales , Ciclina D1/metabolismo , Proteínas de Unión al ADN/metabolismo , Estructuras Embrionarias/anatomía & histología , Estructuras Embrionarias/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Folículo Piloso/anatomía & histología , Proteínas Hedgehog , Inmunohistoquímica , Hibridación in Situ , Factor de Unión 1 al Potenciador Linfoide , Glándulas Mamarias Animales/anatomía & histología , Ratones , Ratones Transgénicos , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Transactivadores/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA