Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 3 de 3
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Oncogene ; 35(11): 1461-7, 2016 Mar 17.
Article de Anglais | MEDLINE | ID: mdl-26096929

RÉSUMÉ

In the normal mammary gland, the basal epithelium is known to be bipotent and can generate either basal or luminal cells, whereas the luminal epithelium has not been demonstrated to contribute to the basal compartment in an intact and normally developed mammary gland. It is not clear whether cellular heterogeneity within a breast tumor results from transformation of bipotent basal cells or from transformation and subsequent basal conversion of the more differentiated luminal cells. Here we used a retroviral vector to express an oncogene specifically in a small number of the mammary luminal epithelial cells and tested their potential to produce basal cells during tumorigenesis. This in-vivo lineage-tracing work demonstrates that luminal cells are capable of producing basal cells on activation of either polyoma middle T antigen or ErbB2 signaling. These findings reveal the plasticity of the luminal compartment during tumorigenesis and provide an explanation for cellular heterogeneity within a cancer.


Sujet(s)
Tumeurs du sein/anatomopathologie , Plasticité cellulaire/physiologie , Cellules épithéliales/cytologie , Glandes mammaires humaines/cytologie , Antigènes des virus oncogènes/métabolisme , Lignage cellulaire/physiologie , Prolifération cellulaire/physiologie , Transformation cellulaire néoplasique/anatomopathologie , Femelle , Humains , Récepteur ErbB-2/métabolisme , Transduction du signal/physiologie
2.
Mol Cell Endocrinol ; 382(1): 560-569, 2014 Jan 25.
Article de Anglais | MEDLINE | ID: mdl-23541951

RÉSUMÉ

The mammary gland is a unique organ that undergoes extensive and profound changes during puberty, menstruation, pregnancy, lactation and involution. The changes that take place during puberty involve large-scale proliferation and invasion of the fat-pad. During pregnancy and lactation, the mammary cells are exposed to signaling pathways that inhibit apoptosis, induce proliferation and invoke terminal differentiation. Finally, during involution the mammary gland is exposed to milk stasis, programmed cell death and stromal reorganization to clear the differentiated milk-producing cells. Not surprisingly, the signaling pathways responsible for bringing about these changes in breast cells are often subverted during the process of tumorigenesis. The STAT family of proteins is involved in every stage of mammary gland development, and is also frequently implicated in breast tumorigenesis. While the roles of STAT3 and STAT5 during mammary gland development and tumorigenesis are well studied, others members, e.g. STAT1 and STAT6, have only recently been observed to play a role in mammary gland biology. Continued investigation into the STAT protein network in the mammary gland will likely yield new biomarkers and risk factors for breast cancer, and may also lead to novel prophylactic or therapeutic strategies against breast cancer.


Sujet(s)
Carcinogenèse/anatomopathologie , Différenciation cellulaire , Glandes mammaires animales/anatomopathologie , Glandes mammaires humaines/anatomopathologie , Facteurs de transcription STAT/métabolisme , Transduction du signal , Animaux , Carcinogenèse/métabolisme , Survie cellulaire , Femelle , Humains , Glandes mammaires animales/métabolisme , Glandes mammaires humaines/métabolisme
3.
Oncogene ; 33(50): 5729-39, 2014 Dec 11.
Article de Anglais | MEDLINE | ID: mdl-24317513

RÉSUMÉ

Pregnancy-associated breast cancers (PABCs) are tumors diagnosed during pregnancy or up to 5 years following parturition, and are usually high-grade, connective tissue-rich, and estrogen receptor (ER)/progesterone receptor-negative. Little is known about the cellular origin of PABCs or the mechanisms by which PABCs are initiated. Using the RCAS retrovirus to deliver the ErbB2 oncogene into the mammary epithelium of our previously reported MMTV-tva transgenic mice, we detected high-grade, poorly differentiated, stroma-rich and ER-negative tumors during pregnancy and lactation. These high-grade and stroma-rich tumors were less frequent in involuted mice or in age-matched nulliparous mice. More importantly, by generating a WAP-tva transgenic line for expression of ErbB2 selectively in WAP(+) mammary alveolar cells, we found that tumors had similar morphological phenotypes (high grade, poorly differentiated, stroma-rich and ER-negative), irrespective of the time since pregnancy and even in the absence of pregnancy. These data suggest that PABCs arise preferentially from an alveolar cell population that expands during pregnancy and lactation. This somatic mouse model may also be useful for preclinical testing of new prophylactic and therapeutic strategies against PABC.


Sujet(s)
Glandes mammaires animales/anatomopathologie , Tumeurs expérimentales de la mamelle/anatomopathologie , Cellules souches tumorales/anatomopathologie , Complications tumorales de la grossesse/anatomopathologie , Animaux , Transformation cellulaire virale , Femelle , Humains , Souris , Souris transgéniques , Grading des tumeurs , Cellules souches tumorales/physiologie , Phénotype , Grossesse
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE