Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Development ; 144(7): 1317-1327, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28275010

ABSTRACT

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.


Subject(s)
Epithelium/metabolism , Mammary Glands, Animal/cytology , Mammary Glands, Animal/growth & development , Morphogenesis , Patched-1 Receptor/metabolism , Animals , Bone Marrow Transplantation , Cell Proliferation/drug effects , Cell Shape/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelium/drug effects , Estrogens/pharmacology , Female , Hedgehog Proteins/metabolism , Integrases/metabolism , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/transplantation , Mice , Models, Biological , Morphogenesis/drug effects , Mutation/genetics , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Progesterone/pharmacology , Signal Transduction/drug effects , Smoothened Receptor/metabolism
3.
J Mammary Gland Biol Neoplasia ; 23(4): 237-248, 2018 12.
Article in English | MEDLINE | ID: mdl-30338425

ABSTRACT

Ductal carcinoma in situ (DCIS) is a non-obligate precursor to most types of invasive breast cancer (IBC). Although it is estimated only one third of untreated patients with DCIS will progress to IBC, standard of care for treatment is surgery and radiation. This therapeutic approach combined with a lack of reliable biomarker panels to predict DCIS progression is a major clinical problem. DCIS shares the same molecular subtypes as IBC including estrogen receptor (ER) and progesterone receptor (PR) positive luminal subtypes, which encompass the majority (60-70%) of DCIS. Compared to the established roles of ER and PR in luminal IBC, much less is known about the roles and mechanism of action of estrogen (E2) and progesterone (P4) and their cognate receptors in the development and progression of DCIS. This is an underexplored area of research due in part to a paucity of suitable experimental models of ER+/PR + DCIS. This review summarizes information from clinical and observational studies on steroid hormones as breast cancer risk factors and ER and PR as biomarkers in DCIS. Lastly, we discuss emerging experimental models of ER+/PR+ DCIS.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Animals , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Breast/pathology , Breast Neoplasms/diagnosis , Breast Neoplasms/therapy , Carcinoma, Intraductal, Noninfiltrating/diagnosis , Carcinoma, Intraductal, Noninfiltrating/therapy , Clinical Trials as Topic , Disease Models, Animal , Disease Progression , Estrogens/metabolism , Female , Humans , Neoplasm Invasiveness/pathology , Observational Studies as Topic , Predictive Value of Tests , Progesterone/metabolism , Risk Factors
4.
Dev Biol ; 352(1): 116-27, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21276786

ABSTRACT

The Hedgehog (Hh) signaling network is critical for patterning and organogenesis in mammals, and has been implicated in a variety of cancers. Smoothened (Smo), the gene encoding the principal signal transducer, is overexpressed frequently in breast cancer, and constitutive activation in MMTV-SmoM2 transgenic mice caused alterations in mammary gland morphology, increased proliferation, and changes in stem/progenitor cell number. Both in transgenic mice and in clinical specimens, proliferative cells did not usually express detectable Smo, suggesting the hypothesis that Smo functioned in a non-cell autonomous manner to stimulate proliferation. Here, we employed a genetically tagged mouse model carrying a Cre-recombinase-dependent conditional allele of constitutively active Smo (SmoM2) to test this hypothesis. MMTV-Cre- or adenoviral-Cre-mediated SmoM2 expression in the luminal epithelium, but not in the myoepithelium, was required for the hyper-proliferative phenotypes. High levels of proliferation were observed in cells adjacent or in close-proximity to Smo expressing cells demonstrating that SmoM2 expressing cells were stimulating proliferation via a paracrine or juxtacrine mechanism. In contrast, Smo expression altered luminal cell differentiation in a cell-autonomous manner. SmoM2 expressing cells, purified by fluorescence activated cell sorting (FACS) via the genetic fluorescent tag, expressed high levels of Ptch2, Gli1, Gli2, Jag2 and Dll-1, and lower levels of Notch4 and Hes6, in comparison to wildtype cells. These studies provide insight into the mechanism of Smo activation in the mammary gland and its possible roles in breast tumorigenesis. In addition, these results also have potential implications for the interpretation of proliferative phenotypes commonly observed in other organs as a consequence of hedgehog signaling activation.


Subject(s)
Cell Differentiation , Epithelial Cells/pathology , Mammary Glands, Animal/pathology , Paracrine Communication , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Communication , Cell Proliferation , Collagen/metabolism , Down-Regulation , Epithelial Cells/metabolism , Female , Hedgehog Proteins/metabolism , Hyperplasia , Integrases/metabolism , Macrophages/cytology , Macrophages/metabolism , Mammary Glands, Animal/metabolism , Mice , Mice, Transgenic , Phenotype , Receptors, Notch/metabolism , Signal Transduction , Smoothened Receptor
5.
Stem Cells ; 28(3): 535-44, 2010 Mar 31.
Article in English | MEDLINE | ID: mdl-20054865

ABSTRACT

The bZIP transcription factor C/EBP beta is important for mammary gland development and its expression is deregulated in human breast cancer. To determine whether C/EBP beta regulates mammary stem cells (MaSCs), we employed two different knockout strategies. Using both a germline and a conditional knockout strategy, we demonstrate that mammosphere formation was significantly decreased in C/EBP beta-deficient mammary epithelial cells (MECs). Functional limiting dilution transplantation assays indicated that the repopulating ability of C/EBP beta-deleted MECs was severely impaired. Serial transplantation experiments demonstrated that C/EBP beta deletion resulted in decreased outgrowth potential and premature MaSC senescence. In accord, fluorescence-activated cell sorting analysis demonstrated that C/EBP beta-null MECs contained fewer MaSCs, the loss of luminal progenitors and an increase in differentiated luminal cells as compared with wild-type. Gene profiling of C/EBP beta-null stem cells revealed an alteration in cell fate specification, exemplified by the expression of basal markers in the luminal compartment. Thus, C/EBP beta is a critical regulator of both MaSC repopulation activity and luminal cell lineage commitment. These findings have critical implications for understanding both stem cell biology and the etiology of different breast cancer subtypes.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/genetics , Cell Differentiation/physiology , Cell Lineage/genetics , Epithelial Cells/metabolism , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Stem Cells/metabolism , Animals , Biomarkers/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/physiopathology , Cell Compartmentation/genetics , Cell Death/genetics , Cell Proliferation , Cellular Senescence/genetics , Epithelial Cells/cytology , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental/genetics , Mammary Glands, Animal/cytology , Mice , Mice, Knockout , Organogenesis/genetics , Stem Cells/cytology
6.
J Mammary Gland Biol Neoplasia ; 14(4): 411-5, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19936989

ABSTRACT

Whole mount preparations of mouse mammary glands are useful for evaluating overall changes in growth and morphology, and are essential for detecting and evaluating focal or regionally-localized phenotypes that would be difficult to detect or analyze using other techniques. We present three newly developed methods for preparing whole mounts of mammary glands from genetically-engineered mice expressing fluorescent proteins, as well as using either neutral red or a variety of fluorescent dyes. Unlike traditional hematoxylin- or carmine-stained preparations, neutral red-stained and some fluorescent preparations can be used for several common downstream analyses.


Subject(s)
Mammary Glands, Animal/anatomy & histology , Tissue and Organ Harvesting/methods , Animals , Female , Fluorescent Dyes , Mice , Mice, Transgenic , Microscopy, Fluorescence , Neutral Red , Staining and Labeling
7.
Sci Signal ; 8(394): ra92, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26373672

ABSTRACT

Hedgehog (Hh) signaling is critical for organogenesis, tissue homeostasis, and stem cell maintenance. The gene encoding Smoothened (SMO), the primary effector of Hh signaling, is expressed aberrantly in human breast cancer, as well as in other cancers. In mice that express a constitutively active form of SMO that does not require Hh stimulation in mammary glands, the cells near the transgenic cells proliferate and participate in hyperplasia formation. Although SMO is a seven-transmembrane receptor like G protein-coupled receptors (GPCRs), SMO-mediated activation of the Gli family of transcription factors is not known to involve G proteins. However, data from Drosophila and mammalian cell lines indicate that SMO functions as a GPCR that couples to heterotrimeric G proteins of the pertussis toxin (PTX)-sensitive Gαi class. Using genetically modified mice, we demonstrated that SMO signaling through G proteins occurred in the mammary gland in vivo. SMO-induced stimulation of proliferation was PTX-sensitive and required Gαi2, but not Gαi1, Gαi3, or activation of Gli1 or Gli2. Our findings show that activated SMO functions as a GPCR to stimulate proliferation in vivo, a finding that may have clinical importance because most SMO-targeted agents have been selected based largely on their ability to block Gli-mediated transcription.


Subject(s)
Cell Proliferation/physiology , Epithelial Cells/metabolism , GTP-Binding Protein alpha Subunit, Gi2/metabolism , Mammary Glands, Animal/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Animals , Drosophila , Epithelial Cells/cytology , Female , GTP-Binding Protein alpha Subunit, Gi2/genetics , Humans , Mammary Glands, Animal/cytology , Mice , Mice, Transgenic , Receptors, G-Protein-Coupled/genetics , Smoothened Receptor
8.
Curr Drug Targets ; 11(9): 1103-11, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20545610

ABSTRACT

The hedgehog signal transduction network is a critical regulator of metazoan development. Inappropriate activation of this network is implicated in several different cancers, including breast. Genetic evidence in mice as well as molecular biological studies in human cells clearly indicate that activated signaling can lead to mammary hyperplasia and, in some cases, tumor formation. However, the exact role(s) activated hedgehog signaling plays in the development or progression of breast cancer also remain unclear. In this review, we have discussed recent data regarding the mechanism(s) by which the hedgehog network may signal in the mammary gland, as well as the data implicating activated signaling as a contributing factor to breast cancer development. Finally, we provide a brief update on the available hedgehog signaling inhibitors with respect to ongoing clinical trials, some of which will include locally advanced or metastatic breast cancers. Given the growing intensity with which the hedgehog signaling network is being studied in the normal and neoplastic mammary gland, a more complete understanding of this network should allow more effective targeting of its activities in breast cancer treatment or prevention.


Subject(s)
Breast Neoplasms/metabolism , Hedgehog Proteins/metabolism , Mammary Glands, Human/metabolism , Signal Transduction , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Hedgehog Proteins/genetics , Humans , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Glands, Human/growth & development , Mammary Glands, Human/pathology , Mice , Mice, Knockout , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL