Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Nature ; 584(7822): 608-613, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32848220

RESUMO

Glandular epithelia, including the mammary and prostate glands, are composed of basal cells (BCs) and luminal cells (LCs)1,2. Many glandular epithelia develop from multipotent basal stem cells (BSCs) that are replaced in adult life by distinct pools of unipotent stem cells1,3-8. However, adult unipotent BSCs can reactivate multipotency under regenerative conditions and upon oncogene expression3,9-13. This suggests that an active mechanism restricts BSC multipotency under normal physiological conditions, although the nature of this mechanism is unknown. Here we show that the ablation of LCs reactivates the multipotency of BSCs from multiple epithelia both in vivo in mice and in vitro in organoids. Bulk and single-cell RNA sequencing revealed that, after LC ablation, BSCs activate a hybrid basal and luminal cell differentiation program before giving rise to LCs-reminiscent of the genetic program that regulates multipotency during embryonic development7. By predicting ligand-receptor pairs from single-cell data14, we find that TNF-which is secreted by LCs-restricts BC multipotency under normal physiological conditions. By contrast, the Notch, Wnt and EGFR pathways were activated in BSCs and their progeny after LC ablation; blocking these pathways, or stimulating the TNF pathway, inhibited regeneration-induced BC multipotency. Our study demonstrates that heterotypic communication between LCs and BCs is essential to maintain lineage fidelity in glandular epithelial stem cells.


Assuntos
Comunicação Celular , Células Epiteliais/citologia , Células-Tronco Multipotentes/citologia , Animais , Linhagem da Célula , Células Epiteliais/metabolismo , Receptores ErbB/metabolismo , Feminino , Homeostase , Humanos , Masculino , Glândulas Mamárias Animais/citologia , Camundongos , Células-Tronco Multipotentes/metabolismo , Organoides/citologia , Próstata/citologia , RNA Mensageiro/genética , RNA-Seq , Receptores Notch/metabolismo , Glândulas Salivares/citologia , Análise de Célula Única , Pele/citologia , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Wnt/metabolismo
2.
Nature ; 525(7567): 119-23, 2015 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-26266985

RESUMO

Breast cancer is the most frequent cancer in women and consists of heterogeneous types of tumours that are classified into different histological and molecular subtypes. PIK3CA and P53 (also known as TP53) are the two most frequently mutated genes and are associated with different types of human breast cancers. The cellular origin and the mechanisms leading to PIK3CA-induced tumour heterogeneity remain unknown. Here we used a genetic approach in mice to define the cellular origin of Pik3ca-derived tumours and the impact of mutations in this gene on tumour heterogeneity. Surprisingly, oncogenic Pik3ca(H1047R) mutant expression at physiological levels in basal cells using keratin (K)5-CreER(T2) mice induced the formation of luminal oestrogen receptor (ER)-positive/progesterone receptor (PR)-positive tumours, while its expression in luminal cells using K8-CReER(T2) mice gave rise to luminal ER(+)PR(+) tumours or basal-like ER(-)PR(-) tumours. Concomitant deletion of p53 and expression of Pik3ca(H1047R) accelerated tumour development and induced more aggressive mammary tumours. Interestingly, expression of Pik3ca(H1047R) in unipotent basal cells gave rise to luminal-like cells, while its expression in unipotent luminal cells gave rise to basal-like cells before progressing into invasive tumours. Transcriptional profiling of cells that underwent cell fate transition upon Pik3ca(H1047R) expression in unipotent progenitors demonstrated a profound oncogene-induced reprogramming of these newly formed cells and identified gene signatures characteristic of the different cell fate switches that occur upon Pik3ca(H1047R) expression in basal and luminal cells, which correlated with the cell of origin, tumour type and different clinical outcomes. Altogether our study identifies the cellular origin of Pik3ca-induced tumours and reveals that oncogenic Pik3ca(H1047R) activates a multipotent genetic program in normally lineage-restricted populations at the early stage of tumour initiation, setting the stage for future intratumoural heterogeneity. These results have important implications for our understanding of the mechanisms controlling tumour heterogeneity and the development of new strategies to block PIK3CA breast cancer initiation.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/patologia , Fosfatidilinositol 3-Quinases/genética , Animais , Neoplasias da Mama/metabolismo , Diferenciação Celular/genética , Divisão Celular , Linhagem da Célula , Transformação Celular Neoplásica , Classe I de Fosfatidilinositol 3-Quinases , Feminino , Genes p53/genética , Humanos , Neoplasias Mamárias Animais/metabolismo , Camundongos , Mutação/genética , Invasividade Neoplásica/genética , Fenótipo , Fosfatidilinositol 3-Quinases/metabolismo , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo
3.
Nature ; 479(7372): 189-93, 2011 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-21983963

RESUMO

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.


Assuntos
Linhagem da Célula , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/crescimento & desenvolvimento , Células-Tronco/citologia , Envelhecimento , Animais , Diferenciação Celular , Transplante de Células , Epitélio , Feminino , Homeostase , Lactação/fisiologia , Glândulas Mamárias Animais/fisiologia , Glândulas Mamárias Animais/transplante , Camundongos , Células-Tronco Multipotentes/citologia , Gravidez , Células-Tronco/metabolismo
4.
Heliyon ; 9(7): e17842, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37456014

RESUMO

The mammary gland (MG) is composed of three main epithelial lineages, the basal cells (BC), the estrogen receptor (ER) positive luminal cells (ER+ LC), and the ER negative LC (ER- LC). Defining the cell identity of each lineage and how it is modulated throughout the different stages of life is important to understand how these cells function and communicate throughout life. Here, we used transgenic mice specifically labelling ER+ LC combined to cell surface markers to isolate with high purity the 3 distinct cell lineages of the mammary gland and defined their expression profiles and chromatin landscapes by performing bulk RNAseq and ATACseq of these isolated populations in puberty, adulthood and mid-pregnancy. Our analysis identified conserved genes, ligands and transcription factor (TF) associated with a specific lineage throughout life as well as genes, ligands and TFs specific for a particular stage of the MG. In summary, our study identified genes and TF network associated with the identity, function and cell-cell communication of the different epithelial lineages of the MG at different stages of life.

5.
Cell Rep ; 20(7): 1525-1532, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28813665

RESUMO

The mammary gland (MG) is composed of different cell lineages, including the basal and the luminal cells (LCs) that are maintained by distinct stem cell (SC) populations. LCs can be subdivided into estrogen receptor (ER)+ and ER- cells. LCs act as the cancer cell of origin in different types of mammary tumors. It remains unclear whether the heterogeneity found in luminal-derived mammary tumors arises from a pre-existing heterogeneity within LCs. To investigate LC heterogeneity, we used lineage tracing to assess whether the ER+ lineage is maintained by multipotent SCs or by lineage-restricted SCs. To this end, we generated doxycycline-inducible ER-rtTA mice that allowed us to perform genetic lineage tracing of ER+ LCs and study their fate and long-term maintenance. Our results show that ER+ cells are maintained by lineage-restricted SCs that exclusively contribute to the expansion of the ER+ lineage during puberty and their maintenance during adult life.


Assuntos
Rastreamento de Células/métodos , Células Epiteliais/citologia , Homeostase/genética , Glândulas Mamárias Animais/citologia , Receptores de Estrogênio/genética , Células-Tronco/citologia , Animais , Diferenciação Celular , Linhagem da Célula , Doxiciclina/farmacologia , Células Epiteliais/metabolismo , Células Epiteliais/transplante , Feminino , Efeito Fundador , Expressão Gênica/efeitos dos fármacos , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Transgênicos , Receptores de Estrogênio/metabolismo , Regeneração/genética , Transplante de Células-Tronco , Células-Tronco/metabolismo
6.
Nat Cell Biol ; 14(11): 1131-8, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23064263

RESUMO

The prostate is a glandular epithelium composed of basal, luminal and neuroendocrine cells that originate from the urogenital sinus during embryonic development. After birth, the prostate keeps developing until the end of puberty. Here, we used inducible genetic lineage tracing experiments in mice to investigate the cellular hierarchy that governs prostate postnatal development. We found that prostate postnatal development is mediated by basal multipotent stem cells that differentiate into basal, luminal and neuroendocrine cells, as well as by unipotent basal and luminal progenitors. Clonal analysis of basal cells revealed the existence of bipotent and unipotent basal progenitors as well as basal cells already committed to the luminal lineage with intermediate cells co-expressing basal and luminal markers associated with this commitment step. The existence of multipotent basal progenitors during prostate postnatal development contrasts with the distinct pools of unipotent basal and luminal stem cells that mediate adult prostate regeneration. Our results uncover the cellular hierarchy acting during prostate development and will be instrumental in defining the cellular origin and the mechanisms underlying prostate cancer initiation.


Assuntos
Células-Tronco Multipotentes/citologia , Próstata/citologia , Células-Tronco/citologia , Animais , Diferenciação Celular/fisiologia , Proliferação de Células , Masculino , Camundongos , Células-Tronco Multipotentes/metabolismo , Próstata/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Células-Tronco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA