Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
EMBO J ; 43(12): 2308-2336, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38760574

RESUMO

How cells coordinate morphogenetic cues and fate specification during development remains a fundamental question in organogenesis. The mammary gland arises from multipotent stem cells (MaSCs), which are progressively replaced by unipotent progenitors by birth. However, the lack of specific markers for early fate specification has prevented the delineation of the features and spatial localization of MaSC-derived lineage-committed progenitors. Here, using single-cell RNA sequencing from E13.5 to birth, we produced an atlas of matched mouse mammary epithelium and mesenchyme and reconstructed the differentiation trajectories of MaSCs toward basal and luminal fate. We show that murine MaSCs exhibit lineage commitment just prior to the first sprouting events of mammary branching morphogenesis at E15.5. We identify early molecular markers for committed and multipotent MaSCs and define their spatial distribution within the developing tissue. Furthermore, we show that the mammary embryonic mesenchyme is composed of two spatially restricted cell populations, and that dermal mesenchyme-produced FGF10 is essential for embryonic mammary branching morphogenesis. Altogether, our data elucidate the spatiotemporal signals underlying lineage specification of multipotent MaSCs, and uncover the signals from mesenchymal cells that guide mammary branching morphogenesis.


Assuntos
Linhagem da Célula , Células Epiteliais , Glândulas Mamárias Animais , Células-Tronco Mesenquimais , Animais , Camundongos , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/embriologia , Glândulas Mamárias Animais/metabolismo , Feminino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Diferenciação Celular , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Fator 10 de Crescimento de Fibroblastos/metabolismo , Fator 10 de Crescimento de Fibroblastos/genética , Morfogênese , Análise de Célula Única , Mesoderma/citologia , Mesoderma/metabolismo , Mesoderma/embriologia
2.
Nat Comput Sci ; 4(2): 128-143, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38374363

RESUMO

Identifying true DNA cellular barcodes among polymerase chain reaction and sequencing errors is challenging. Current tools are restricted in the diversity of barcode types supported or the analysis strategies implemented. As such, there is a need for more versatile and efficient tools for barcode extraction, as well as for tools to investigate which factors impact barcode detection and which filtering strategies to best apply. Here we introduce the package CellBarcode and its barcode simulation kit, CellBarcodeSim, that allows efficient and versatile barcode extraction and filtering for a range of barcode types from bulk or single-cell sequencing data using a variety of filtering strategies. Using the barcode simulation kit and biological data, we explore the technical and biological factors influencing barcode identification and provide a decision tree on how to optimize barcode identification for different barcode settings. We believe that CellBarcode and CellBarcodeSim have the capability to enhance the reproducibility and interpretation of barcode results across studies.


Assuntos
Código de Barras de DNA Taxonômico , DNA , Reprodutibilidade dos Testes , Análise de Sequência de DNA/métodos , Código de Barras de DNA Taxonômico/métodos , DNA/genética , Reação em Cadeia da Polimerase
3.
Elife ; 112022 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-35543624

RESUMO

Tumours are complex ecosystems composed of different types of cells that communicate and influence each other. While the critical role of stromal cells in affecting tumour growth is well established, the impact of mutant cancer cells on healthy surrounding tissues remains poorly defined. Here, using mouse intestinal organoids, we uncover a paracrine mechanism by which intestinal cancer cells reactivate foetal and regenerative YAP-associated transcriptional programmes in neighbouring wildtype epithelial cells, rendering them adapted to thrive in the tumour context. We identify the glycoprotein thrombospondin-1 (THBS1) as the essential factor that mediates non-cell-autonomous morphological and transcriptional responses. Importantly, Thbs1 is associated with bad prognosis in several human cancers. This study reveals the THBS1-YAP axis as the mechanistic link mediating paracrine interactions between epithelial cells in intestinal tumours.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Neoplasias , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Ecossistema , Células Epiteliais/metabolismo , Camundongos , Transdução de Sinais , Fatores de Transcrição/metabolismo
4.
Cell Rep ; 38(10): 110461, 2022 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-35263603

RESUMO

Real-time in vivo imaging provides an essential window into the spatiotemporal cellular events contributing to tissue development and pathology. By coupling longitudinal intravital imaging with genetic lineage tracing, here we capture the earliest cellular events arising in response to active Wnt/ß-catenin signaling and the ensuing impact on the organization and differentiation of the mammary epithelium. This enables us to interrogate how Wnt/ß-catenin regulates the dynamics of distinct subpopulations of mammary epithelial cells in vivo and in real time. We show that ß-catenin stabilization, when targeted to either the mammary luminal or basal epithelial lineage, leads to cellular rearrangements that precipitate the formation of hyperplastic lesions that undergo squamous transdifferentiation. These results enhance our understanding of the earliest stages of hyperplastic lesion formation in vivo and reveal that, in mammary neoplastic development, ß-catenin activation dictates a hair follicle/epidermal differentiation program independently of the targeted cell of origin.


Assuntos
Glândulas Mamárias Animais , beta Catenina , Animais , Células Epiteliais/metabolismo , Epitélio/metabolismo , Hiperplasia/patologia , Glândulas Mamárias Animais/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo
5.
Sci Rep ; 9(1): 888, 2019 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-30696875

RESUMO

Colon tumours are hierarchically organized and contain multipotent self-renewing cells, called Cancer Stem Cells (CSCs). We have previously shown that the Notch1 receptor is expressed in Intestinal Stem Cells (ISCs); given the critical role played by Notch signalling in promoting intestinal tumourigenesis, we explored Notch1 expression in tumours. Combining lineage tracing in two tumour models with transcriptomic analyses, we found that Notch1+ tumour cells are undifferentiated, proliferative and capable of indefinite self-renewal and of generating a heterogeneous clonal progeny. Molecularly, the transcriptional signature of Notch1+ tumour cells highly correlates with ISCs, suggestive of their origin from normal crypt cells. Surprisingly, Notch1+ expression labels a subset of CSCs that shows reduced levels of Lgr5, a reported CSCs marker. The existence of distinct stem cell populations within intestinal tumours highlights the necessity of better understanding their hierarchy and behaviour, to identify the correct cellular targets for therapy.


Assuntos
Linhagem da Célula/fisiologia , Células-Tronco Neoplásicas/metabolismo , Receptor Notch1/genética , Animais , Biomarcadores/metabolismo , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Autorrenovação Celular/fisiologia , Transformação Celular Neoplásica/metabolismo , Neoplasias do Colo/patologia , Perfilação da Expressão Gênica/métodos , Humanos , Mucosa Intestinal/metabolismo , Neoplasias Intestinais/patologia , Intestinos/patologia , Intestinos/fisiologia , Camundongos , Camundongos Transgênicos , Receptor Notch1/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais
6.
Nat Cell Biol ; 20(6): 677-687, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29784917

RESUMO

Recent lineage tracing studies have revealed that mammary gland homeostasis relies on unipotent stem cells. However, whether and when lineage restriction occurs during embryonic mammary development, and which signals orchestrate cell fate specification, remain unknown. Using a combination of in vivo clonal analysis with whole mount immunofluorescence and mathematical modelling of clonal dynamics, we found that embryonic multipotent mammary cells become lineage-restricted surprisingly early in development, with evidence for unipotency as early as E12.5 and no statistically discernable bipotency after E15.5. To gain insights into the mechanisms governing the switch from multipotency to unipotency, we used gain-of-function Notch1 mice and demonstrated that Notch activation cell autonomously dictates luminal cell fate specification to both embryonic and basally committed mammary cells. These functional studies have important implications for understanding the signals underlying cell plasticity and serve to clarify how reactivation of embryonic programs in adult cells can lead to cancer.


Assuntos
Diferenciação Celular , Linhagem da Célula , Plasticidade Celular , Células Epiteliais/metabolismo , Glândulas Mamárias Animais/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo , Receptor Notch1/metabolismo , Células-Tronco Adultas/metabolismo , Células-Tronco Adultas/patologia , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Feminino , Imunofluorescência , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Glândulas Mamárias Animais/embriologia , Camundongos , Camundongos Transgênicos , Modelos Genéticos , Morfogênese , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Fenótipo , Receptor Notch1/genética , Transdução de Sinais , Análise de Célula Única , Fatores de Tempo
7.
PLoS Biol ; 13(2): e1002069, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25688859

RESUMO

The hierarchical relationships between stem cells and progenitors that guide mammary gland morphogenesis are still poorly defined. While multipotent basal stem cells have been found within the myoepithelial compartment, the in vivo lineage potential of luminal progenitors is unclear. Here we used the expression of the Notch1 receptor, previously implicated in mammary gland development and tumorigenesis, to elucidate the hierarchical organization of mammary stem/progenitor cells by lineage tracing. We found that Notch1 expression identifies multipotent stem cells in the embryonic mammary bud, which progressively restrict their lineage potential during mammary ductal morphogenesis to exclusively generate an ERαneg luminal lineage postnatally. Importantly, our results show that Notch1-labelled cells represent the alveolar progenitors that expand during pregnancy and survive multiple successive involutions. This study reveals that postnatal luminal epithelial cells derive from distinct self-sustained lineages that may represent the cells of origin of different breast cancer subtypes.


Assuntos
Linhagem da Célula/genética , Células Epiteliais/citologia , Glândulas Mamárias Animais/citologia , Células-Tronco Multipotentes/citologia , Organogênese/genética , Animais , Diferenciação Celular , Rastreamento de Células , Células Epiteliais/metabolismo , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Introdução de Genes , Integrases/genética , Integrases/metabolismo , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/metabolismo , Camundongos , Camundongos Transgênicos , Células-Tronco Multipotentes/metabolismo , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Fenótipo , Gravidez , Receptor Notch1/genética , Receptor Notch1/metabolismo
8.
Stem Cell Res ; 13(3 Pt A): 355-66, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25290188

RESUMO

Deletion studies confirm Wnt, Notch and Myb transcriptional pathway engagement in intestinal tumorigenesis. Nevertheless, their contrasting and combined roles when activated have not been elucidated. This is important as these pathways are not ablated but rather are aberrantly activated during carcinogenesis. Using ApcMin/+ mice as a source of organoids we documented their transition, on a clone-by-clone basis, to cyst-like spheres with constitutively activated Wnt pathway, increased self-renewal and growth and reduced differentiation. We then looked at this transition when Myb and/or Notch1 are activated. Activated Notch promoted cyst-like organoids. Conversely growth and propagation of cyst-like, but not normal organoids were Notch-independent. Activated Myb promoted normal, but not cyst-like organoids. Interestingly the Wnt, Notch and Myb pathways were all involved in regulating the expression of the intestinal stem cell (ISC) gene Lgr5 in organoids, while ISC gene and Notch target Olfm4 was dominantly repressed by Wnt. These findings parallel mouse intestinal adenoma formation where Notch promoted the initiation, but not growth, of Wnt-driven Olfm4-repressed colon tumors. Also Myb was essential for colon tumor initiation and collateral mouse pathologies. These data reveal the complex interplay and hierarchy of transcriptional networks that operate in ISCs and uncover a shift in pathway-dependencies during tumor initiation.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Proteínas Proto-Oncogênicas c-myb/metabolismo , Receptores Notch/metabolismo , Células-Tronco/metabolismo , Proteínas Wnt/metabolismo , Adenoma/metabolismo , Adenoma/mortalidade , Adenoma/patologia , Animais , Carcinogênese , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Mucosa Intestinal/metabolismo , Intestinos/citologia , Estimativa de Kaplan-Meier , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Organoides/citologia , Organoides/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores Notch/genética , Transdução de Sinais , Células-Tronco/citologia
9.
J Cell Biol ; 203(1): 47-56, 2013 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-24100291

RESUMO

The identity of mammary stem and progenitor cells remains poorly understood, mainly as a result of the lack of robust markers. The Notch signaling pathway has been implicated in mammary gland development as well as in tumorigenesis in this tissue. Elevated expression of the Notch3 receptor has been correlated to the highly aggressive "triple negative" human breast cancer. However, the specific cells expressing this Notch paralogue in the mammary gland remain unknown. Using a conditionally inducible Notch3-CreERT2(SAT) transgenic mouse, we genetically marked Notch3-expressing cells throughout mammary gland development and followed their lineage in vivo. We demonstrate that Notch3 is expressed in a highly clonogenic and transiently quiescent luminal progenitor population that gives rise to a ductal lineage. These cells are capable of surviving multiple successive pregnancies, suggesting a capacity to self-renew. Our results also uncover a role for the Notch3 receptor in restricting the proliferation and consequent clonal expansion of these cells.


Assuntos
Linhagem da Célula , Proliferação de Células , Glândulas Mamárias Animais/metabolismo , Receptores Notch/metabolismo , Células-Tronco/metabolismo , Animais , Biomarcadores/metabolismo , Sobrevivência Celular , Técnicas de Cocultura , Células Alimentadoras , Feminino , Glândulas Mamárias Animais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células NIH 3T3 , Gravidez , Receptor Notch3 , Receptores Notch/genética , Transdução de Sinais , Fatores de Tempo
10.
PLoS One ; 6(10): e25785, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21991352

RESUMO

The conserved role of Notch signaling in controlling intestinal cell fate specification and homeostasis has been extensively studied. Nevertheless, the precise identity of the cells in which Notch signaling is active and the role of different Notch receptor paralogues in the intestine remain ambiguous, due to the lack of reliable tools to investigate Notch expression and function in vivo. We generated a new series of transgenic mice that allowed us, by lineage analysis, to formally prove that Notch1 and Notch2 are specifically expressed in crypt stem cells. In addition, a novel Notch reporter mouse, Hes1-EmGFP(SAT), demonstrated exclusive Notch activity in crypt stem cells and absorptive progenitors. This roster of knock-in and reporter mice represents a valuable resource to functionally explore the Notch pathway in vivo in virtually all tissues.


Assuntos
Linhagem da Célula , Técnicas de Introdução de Genes , Intestinos/citologia , Receptores Notch/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Células Clonais , Enterócitos/citologia , Enterócitos/metabolismo , Marcação de Genes , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Integrases/metabolismo , Cinética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microvilosidades/metabolismo , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Fatores de Transcrição HES-1 , Transcrição Gênica
11.
Proc Natl Acad Sci U S A ; 106(15): 6309-14, 2009 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-19251639

RESUMO

Notch and Wnt signals play essential roles in intestinal development and homeostasis, yet how they integrate their action to affect intestinal morphogenesis is not understood. We examined the interplay between these two signaling pathways in vivo, by modulating Notch activity in mice carrying either a loss- or a gain-of-function mutation of Wnt signaling. We find that the dramatic proliferative effect that Notch signals have on early intestinal precursors requires normal Wnt signaling, whereas its influence on intestinal differentiation appears independent of Wnt. Analogous experiments in Drosophila demonstrate that the synergistic effects of Notch and Wnt are valid across species. We also demonstrate a striking synergy between Notch and Wnt signals that results in inducing the formation of intestinal adenomas, particularly in the colon, a region rarely affected in available mouse tumor models, but the primary target organ in human patients. These studies thus reveal a previously unknown oncogenic potential of Notch signaling in colorectal tumorigenesis that, significantly, is supported by the analysis of human tumors. Importantly, our experimental evidence raises the possibility that Notch activation might be an essential initial event triggering colorectal cancer.


Assuntos
Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Neoplasias Intestinais/metabolismo , Neoplasias Intestinais/patologia , Receptores Notch/metabolismo , Transdução de Sinais , Proteínas Wnt/metabolismo , Adenoma/genética , Adenoma/metabolismo , Adenoma/patologia , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Diferenciação Celular , Proliferação de Células , Transformação Celular Neoplásica/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Humanos , Neoplasias Intestinais/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Receptores Notch/genética , Taxa de Sobrevida , Fatores de Transcrição TCF/deficiência , Fatores de Transcrição TCF/genética , Fatores de Transcrição TCF/metabolismo , Fator de Transcrição 4 , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
13.
Nature ; 435(7044): 964-8, 2005 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-15959516

RESUMO

The Notch signalling pathway plays a crucial role in specifying cellular fates in metazoan development by regulating communication between adjacent cells. Correlative studies suggested an involvement of Notch in intestinal development. Here, by modulating Notch activity in the mouse intestine, we directly implicate Notch signals in intestinal cell lineage specification. We also show that Notch activation is capable of amplifying the intestinal progenitor pool while inhibiting cell differentiation. We conclude that Notch activity is required for the maintenance of proliferating crypt cells in the intestinal epithelium.


Assuntos
Linhagem da Célula , Mucosa Intestinal/metabolismo , Intestinos/citologia , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo , Animais , Apoptose , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Diferenciação Celular , Proliferação de Células , Proteínas de Ligação a DNA/genética , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Regulação da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Integrases/genética , Integrases/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor Notch1 , Receptores de Superfície Celular/genética , Fatores de Transcrição HES-1 , Fatores de Transcrição/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...