Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Breast Cancer Res ; 26(1): 91, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38835038

RESUMEN

BACKGROUND: The aberrant amplification of mammary luminal progenitors is at the origin of basal-like breast cancers associated with BRCA1 mutations. Integrins mediate cell-matrix adhesion and transmit mechanical and chemical signals that drive epithelial stem cell functions and regulate tumor progression, metastatic reactivation, and resistance to targeted therapies. Consistently, we have recently shown that laminin-binding integrins are essential for the expansion and differentiation of mammary luminal progenitors in physiological conditions. As over-expression of the laminin-binding α6 integrin (Itgα6) is associated with poor prognosis and reduced survival in breast cancer, we here investigate the role of Itgα6 in mammary tumorigenesis. METHODS: We used Blg-Cre; Brca1F/F; Trp53F/F mice, a model that phenocopies human basal-like breast cancer with BRCA1 mutations. We generated mutant mice proficient or deficient in Itgα6 expression and followed tumor formation. Mammary tumors and pretumoral tissues were characterized by immunohistochemistry, flow cytometry, RT-qPCR, Western blotting and organoid cultures. Clonogenicity of luminal progenitors from preneoplastic glands was studied in 3D Matrigel cultures. RESULTS: We show that Itga6 deletion favors activation of p16 cell cycle inhibitor in the preneoplastic tissue. Subsequently, the amplification of luminal progenitors, the cell of origin of Brca1-deficient tumors, is restrained in Itgα6-deficient gland. In addition, the partial EMT program operating in Brca1/p53-deficient epithelium is attenuated in the absence of Itgα6. As a consequence of these events, mammary tumor formation is delayed in Itgα6-deficient mice. After tumor formation, the lack of Itgα6 does not affect tumor growth but rather alters their differentiation, resulting in reduced expression of basal cell markers. CONCLUSIONS: Our data indicate that Itgα6 has a pro-tumorigenic role in Blg-Cre; Brca1F/F; Trp53F/F mice developing basal-like mammary tumors. In particular, we reveal that Itgα6 is required for the luminal progenitor expansion and the aberrant partial EMT program that precedes the formation of BRCA1 deficient tumors.


Asunto(s)
Proteína BRCA1 , Neoplasias de la Mama , Integrina alfa6 , Proteína p53 Supresora de Tumor , Animales , Integrina alfa6/metabolismo , Integrina alfa6/genética , Femenino , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ratones , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Humanos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Proliferación Celular , Células Madre/metabolismo , Eliminación de Gen , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo
2.
EMBO J ; 43(12): 2308-2336, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38760574

RESUMEN

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.


Asunto(s)
Linaje de la Célula , Células Epiteliales , Glándulas Mamarias Animales , Células Madre Mesenquimatosas , Animales , Ratones , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/embriología , Glándulas Mamarias Animales/metabolismo , Femenino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Diferenciación Celular , Células Madre Multipotentes/citología , Células Madre Multipotentes/metabolismo , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Factor 10 de Crecimiento de Fibroblastos/genética , Morfogénesis , Análisis de la Célula Individual , Mesodermo/citología , Mesodermo/metabolismo , Mesodermo/embriología
3.
Nat Comput Sci ; 4(2): 128-143, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38374363

RESUMEN

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.


Asunto(s)
Código de Barras del ADN Taxonómico , ADN , Reproducibilidad de los Resultados , Análisis de Secuencia de ADN/métodos , Código de Barras del ADN Taxonómico/métodos , ADN/genética , Reacción en Cadena de la Polimerasa
4.
Cell Rep ; 38(10): 110461, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35263603

RESUMEN

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.


Asunto(s)
Glándulas Mamarias Animales , beta Catenina , Animales , Células Epiteliales/metabolismo , Epitelio/metabolismo , Hiperplasia/patología , Glándulas Mamarias Animales/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo
5.
Methods Mol Biol ; 2471: 123-139, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35175594

RESUMEN

The orthotopic transplantation assay has provided important insights into mammary development, stem cell function, and tumorigenesis. Technically, it consists in grafting mammary tissue fragments, organoids, mammospheres, or isolated cells into the fat pads of prepubertal mice from which the endogenous epithelium has been surgically removed, thereby allowing growth and differentiation of mammary epithelial cells in their physiological environment. Here, we describe how is conducted transplantation of epithelial fragments and cells isolated from mouse mammary glands, report the various approaches currently used to evaluate the regeneration and self-renewal properties of mammary stem cells, and highlight the strengths and limitations of this in vivo grafting assay.


Asunto(s)
Células Epiteliales , Glándulas Mamarias Animales , Animales , Diferenciación Celular , Células Epiteliales/trasplante , Epitelio/fisiología , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/fisiología , Ratones , Células Madre
6.
Cancer Res ; 81(17): 4529-4544, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34145034

RESUMEN

Cancer stem cells (CSC) are considered responsible for tumor initiation, therapeutic resistance, and metastasis. A comprehensive knowledge of the mechanisms governing the acquisition and maintenance of cancer stemness is crucial for the development of new therapeutic approaches in oncology. E2A basic helix-loop-helix (bHLH) transcription factors are associated with epithelial-mesenchymal transition (EMT) and tumor progression, but knowledge of their functional contributions to cancer biology is still limited. Using a combination of in vivo and in vitro analyses in a novel PyMT-E2A conditional knockout mouse model and derived primary tumor cell lines, we report here an essential role of E2A in stemness, metastasis, and therapeutic resistance in breast cancer. Targeted deletion of E2A in the mammary gland impaired tumor-initiating ability and dedifferentiation potential and severely compromised metastatic competence of PyMT-driven mammary tumors. Mechanistic studies in PyMT-derived cell lines indicated that E2A actions are mediated by the upregulation of Snai1 transcription. Importantly, high E2A and SNAIL1 expression occurred in aggressive human basal-like breast carcinomas, highlighting the relevance of the E2A-Snail1 axis in metastatic breast cancer. In addition, E2A factors contributed to the maintenance of genomic integrity and resistance to PARP inhibitors in PyMT and human triple-negative breast cancer cells. Collectively, these results support the potential for E2A transcription factors as novel targets worthy of translational consideration in breast cancer. SIGNIFICANCE: These findings identify key functions of E2A factors in breast cancer cell stemness, metastasis, and drug resistance, supporting a therapeutic vulnerability to targeting E2A proteins in breast cancer.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neoplasias de la Mama/metabolismo , Resistencia a Antineoplásicos , Adulto , Anciano , Anciano de 80 o más Años , Animales , Neoplasias de la Mama/genética , Carcinogénesis , Diferenciación Celular , Línea Celular Tumoral , Transformación Celular Neoplásica , Simulación por Computador , Transición Epitelial-Mesenquimal , Femenino , Eliminación de Gen , Genoma , Genotipo , Humanos , Masculino , Neoplasias Mamarias Animales , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Metástasis de la Neoplasia , Células Madre Neoplásicas , Ftalazinas/farmacología , Piperazinas/farmacología , Factores de Transcripción de la Familia Snail/metabolismo , Transgenes , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo
7.
Development ; 147(19)2020 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-32895290

RESUMEN

The Golgi-associated RAB GTPases, RAB6A and RAB6A', regulate anterograde and retrograde transport pathways from and to the Golgi. In vitro, RAB6A/A' control several cellular functions including cell division, migration, adhesion and polarity. However, their role remains poorly described in vivo Here, we generated BlgCre; Rab6aF/F mice presenting a specific deletion of Rab6a in the mammary luminal secretory lineage during gestation and lactation. Rab6a loss severely impaired the differentiation, maturation and maintenance of the secretory tissue, compromising lactation. The mutant epithelium displayed a decreased activation of STAT5, a key regulator of the lactogenic process primarily governed by prolactin. Data obtained with a mammary epithelial cell line suggested that defective STAT5 activation might originate from a perturbed transport of the prolactin receptor, altering its membrane expression and signaling cascade. Despite the major functional defects observed upon Rab6a deletion, the polarized organization of the mammary epithelial bilayer was preserved. Altogether, our data reveal a crucial role for RAB6A/A' in the lactogenic function of the mammary gland and suggest that the trafficking pathways controlled by RAB6A/A' depend on cell-type specialization and tissue context.


Asunto(s)
Glándulas Mamarias Humanas/metabolismo , Factor de Transcripción STAT5/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Animales , Western Blotting , Línea Celular , Femenino , Citometría de Flujo , Humanos , Etiquetado Corte-Fin in Situ , Glándulas Mamarias Humanas/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Factor de Transcripción STAT5/genética , Proteínas de Unión al GTP rab/genética
8.
Development ; 147(4)2020 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-31988184

RESUMEN

Integrin dimers α3/ß1, α6/ß1 and α6/ß4 are the mammary epithelial cell receptors for laminins, which are major components of the mammary basement membrane. The roles of specific basement membrane components and their integrin receptors in the regulation of functional gland development have not been analyzed in detail. To investigate the functions of laminin-binding integrins, we obtained mutant mice with mammary luminal cell-specific deficiencies of the α3 and α6 integrin chains generated using the Cre-Lox approach. During pregnancy, mutant mice displayed decreased luminal progenitor activity and retarded lobulo-alveolar development. Mammary glands appeared functional at the onset of lactation in mutant mice; however, myoepithelial cell morphology was markedly altered, suggesting cellular compensation mechanisms involving cytoskeleton reorganization. Notably, lactation was not sustained in mutant females, and the glands underwent precocious involution. Inactivation of the p53 gene rescued the growth defects but did not restore lactogenesis in mutant mice. These results suggest that the p53 pathway is involved in the control of mammary cell proliferation and survival downstream of laminin-binding integrins, and underline an essential role of cell interactions with laminin for lactogenic differentiation.


Asunto(s)
Integrinas/fisiología , Lactancia , Glándulas Mamarias Animales/fisiología , Animales , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Supervivencia Celular , Citoesqueleto/fisiología , Progresión de la Enfermedad , Femenino , Eliminación de Gen , Hormonas/fisiología , Integrina alfa3/fisiología , Integrina alfa6/fisiología , Integrina beta1/fisiología , Integrina beta4/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Ratones Mutantes , Mutación , Células Madre Neoplásicas/citología , Análisis de Secuencia por Matrices de Oligonucleótidos , Ovario/fisiología , Fenotipo , Embarazo , Preñez , Pronóstico , Unión Proteica , Multimerización de Proteína
10.
Stem Cell Reports ; 12(4): 831-844, 2019 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-30905738

RESUMEN

Integrins, which bind laminin, a major component of the mammary basement membrane, are strongly expressed in basal stem cell-enriched populations, but their role in controlling mammary stem cell function remains unclear. We found that stem cell activity, as evaluated in transplantation and mammosphere assays, was reduced in mammary basal cells depleted of laminin receptors containing α3- and α6-integrin subunits. This was accompanied by low MDM2 levels, p53 stabilization, and diminished proliferative capacity. Importantly, disruption of p53 function restored the clonogenicity of α3/α6-integrin-depleted mammary basal stem cells, while inhibition of RHO or myosin II, leading to decreased p53 activity, rescued the mammosphere formation. These data suggest that α3/α6-integrin-mediated adhesion plays an essential role in controlling the proliferative potential of mammary basal stem/progenitor cells through myosin II-mediated regulation of p53 and indicate that laminins might be important components of the mammary stem cell niche.

11.
Breast Cancer Res ; 21(1): 13, 2019 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-30683141

RESUMEN

BACKGROUND: The adult mammary epithelium is composed of basal and luminal cells. The luminal lineage comprises two major cell populations, positive and negative for estrogen and progesterone receptors (ER and PR, respectively), both containing clonogenic progenitor cells. Deregulated ER/PR- luminal progenitor cells are suspected to be at the origin of basal-type triple-negative (TNBC) breast cancers, a subtype frequently associated with loss of P53 function and MET signaling hyperactivation. Using mouse models, we recently reported that p53 restricts luminal progenitor cell amplification whereas paracrine Met activation stimulates their growth and favors a luminal-to-basal switch. Here, we analyzed how these two critical pathways interact to control luminal progenitor function. METHODS: We have (i) established and analyzed the gene expression profile of luminal progenitors isolated by ICAM-1, a robust surface marker we previously identified; (ii) purified luminal progenitors from p53-deficient and p53-proficient mouse mammary epithelium to compare their functional and molecular characteristics; and (iii) analyzed their response to HGF, the major Met ligand, in three-dimensional cultures. RESULTS: We found that luminal progenitors, compared to non-clonogenic luminal cells, overexpress Trp53 and numerous p53 target genes. In vivo, loss of Trp53 induced the expansion of luminal progenitors, affecting expression of several important p53 target genes including those encoding negative regulators of cell cycle progression. Consistently, p53-deficient luminal progenitors displayed increased proliferative and self-renewal activities in culture. However, they did not exhibit perturbed expression of luminal-specific markers and major regulators, such as Hey1, Elf5, and Gata3. Moreover, although expressing Met at higher level than p53-proficient luminal progenitors, p53-deficient luminal progenitors failed to acquire basal-specific features when stimulated by HGF, showing that p53 promotes the plastic behavior of luminal progenitors downstream of Met activation. CONCLUSIONS: Our study reveals a crosstalk between Met- and p53-mediated signaling pathways in the regulation of luminal progenitor function. In particular, it shows that neither p53 loss alone nor p53 loss combined with Met signaling activation caused an early detectable cell fate alteration in luminal progenitors. Conceivably, additional events are required to confer basal-specific characteristics to luminal-derived TNBCs.


Asunto(s)
Plasticidad de la Célula/fisiología , Glándulas Mamarias Animales/citología , Proteínas Proto-Oncogénicas c-met/metabolismo , Células Madre/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Ciclo Celular/fisiología , Células Epiteliales/fisiología , Femenino , Molécula 1 de Adhesión Intercelular/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Cultivo Primario de Células , Transducción de Señal/fisiología , Proteína p53 Supresora de Tumor/genética
12.
Development ; 145(4)2018 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-29361573

RESUMEN

Stem cells (SCs) drive mammary development, giving rise postnatally to an epithelial bilayer composed of luminal and basal myoepithelial cells. Dysregulation of SCs is thought to be at the origin of certain breast cancers; however, the molecular identity of SCs and the factors regulating their function remain poorly defined. We identified the transmembrane protein podoplanin (Pdpn) as a specific marker of the basal compartment, including multipotent SCs, and found Pdpn localized at the basal-luminal interface. Embryonic deletion of Pdpn targeted to basal cells diminished basal and luminal SC activity and affected the expression of several Wnt/ß-catenin signaling components in basal cells. Moreover, Pdpn loss attenuated mammary tumor formation in a mouse model of ß-catenin-induced breast cancer, limiting tumor-initiating cell expansion and promoting molecular features associated with mesenchymal-to-epithelial cell transition. In line with the loss-of-function data, we demonstrated that mechanistically Pdpn enhances Wnt/ß-catenin signaling in mammary basal cells. Overall, this study uncovers a role for Pdpn in mammary SC function and, importantly, identifies Pdpn as a new regulator of Wnt/ß-catenin signaling, a key pathway in mammary development and tumorigenesis.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Células Madre/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Animales Modificados Genéticamente , Western Blotting , Carcinogénesis/metabolismo , Técnicas de Cultivo de Célula , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Vía de Señalización Wnt/genética
13.
J Cell Biol ; 211(4): 737-9, 2015 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-26598611

RESUMEN

In this issue, Tosoni et al. (2015. J. Cell Biol. http://dx.doi.org/10.1083/jcb.201505037) report that cell fate determinant and tumor suppressor Numb imposes asymmetric cell divisions in mammary stem cells by regulating p53. Numb thereby restricts mammary stem cell expansion and controls the proliferation and lineage-specific characteristics of their progeny.


Asunto(s)
Autorrenovación de las Células , Células Epiteliales/fisiología , Proteínas de la Membrana/fisiología , Proteínas del Tejido Nervioso/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Femenino
14.
Elife ; 42015 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-26165517

RESUMEN

HGF/Met signaling has recently been associated with basal-type breast cancers, which are thought to originate from progenitor cells residing in the luminal compartment of the mammary epithelium. We found that ICAM-1 efficiently marks mammary luminal progenitors comprising hormone receptor-positive and receptor-negative cells, presumably ductal and alveolar progenitors. Both cell populations strongly express Met, while HGF is produced by stromal and basal myoepithelial cells. We show that persistent HGF treatment stimulates the clonogenic activity of ICAM1-positive luminal progenitors, controlling their survival and proliferation, and leads to the expression of basal cell characteristics, including stem cell potential. This is accompanied by the induction of Snai1 and Snai2, two major transcription factors triggering epithelial-mesenchymal transition, the repression of the luminal-regulatory genes Elf5 and Hey1, and claudin down-regulation. Our data strongly indicate that paracrine Met signaling can control the function of luminal progenitors and modulate their fate during mammary development and tumorigenesis.


Asunto(s)
Transición Epitelial-Mesenquimal , Factor de Crecimiento de Hepatocito/metabolismo , Comunicación Paracrina , Proteínas Proto-Oncogénicas c-met/metabolismo , Transducción de Señal , Células Madre/fisiología , Animales , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Células Epiteliales/fisiología , Ratones , Células Madre/efectos de los fármacos
15.
Methods Mol Biol ; 1293: 161-72, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26040687

RESUMEN

The transplantation of mammary epithelial cells into the cleared fat pad allows their growth and differentiation in their normal physiological environment. This technique involves the grafting of tissue fragments or isolated cells into the mammary fat pads of prepubertal mice from which the endogenous epithelium has been surgically removed. Such transplantation assays are particularly useful for the analysis of morphogenetic potential and stem cell activity in normal mammary epithelium and breast tumors. We describe here the main steps in the transplantation of epithelial fragments and isolated cells from mouse mammary glands and the various approaches currently used to evaluate the regeneration and self-renewal properties of mammary stem cells.


Asunto(s)
Tejido Adiposo , Trasplante de Células , Células Epiteliales/citología , Glándulas Mamarias Animales/citología , Animales , Separación Celular/métodos , Trasplante de Células/métodos , Femenino , Ratones
16.
Nat Cell Biol ; 16(10): 942-50, 1-7, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25173976

RESUMEN

Contractile myoepithelial cells dominate the basal layer of the mammary epithelium and are considered to be differentiated cells. However, we observe that up to 54% of single basal cells can form colonies when seeded into adherent culture in the presence of agents that disrupt actin-myosin interactions, and on average, 65% of the single-cell-derived basal colonies can repopulate a mammary gland when transplanted in vivo. This indicates that a high proportion of basal myoepithelial cells can give rise to a mammary repopulating unit (MRU). We demonstrate that myoepithelial cells, flow-sorted using two independent myoepithelial-specific reporter strategies, have MRU capacity. Using an inducible lineage-tracing approach we follow the progeny of myoepithelial cells that express α-smooth muscle actin and show that they function as long-lived lineage-restricted stem cells in the virgin state and during pregnancy.


Asunto(s)
Células Epiteliales/citología , Glándulas Mamarias Animales/citología , Miocitos del Músculo Liso/metabolismo , Células Madre/citología , Actinas/metabolismo , Animales , Benzamidas/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Dioxoles/farmacología , Células Epiteliales/metabolismo , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Glándulas Mamarias Animales/metabolismo , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Microscopía Fluorescente , Miocitos del Músculo Liso/citología , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo
17.
Mol Cancer ; 12(1): 132, 2013 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-24171719

RESUMEN

BACKGROUND: Basal-like breast cancer is a heterogeneous disease characterized by the expression of basal cell markers, no estrogen or progesterone receptor expression and a lack of HER2 overexpression. Recent studies have linked activation of the Wnt/ß-catenin pathway, and its downstream target, Myc, to basal-like breast cancer. Transgenic mice K5ΔNßcat previously generated by our team present a constitutive activation of Wnt/ß-catenin signaling in the basal myoepithelial cell layer, resulting in focal mammary hyperplasias that progress to invasive carcinomas. Mammary lesions developed by K5ΔNßcat mice consist essentially of basal epithelial cells that, in contrast to mammary myoepithelium, do not express smooth muscle markers. METHODS: Microarray analysis was used to compare K5ΔNßcat mouse tumors to human breast tumors, mammary cancer cell lines and the tumors developed in other mouse models. Cre-Lox approach was employed to delete Myc from the mammary basal cell layer of K5ΔNßcat mice. Stem cell amplification in K5ΔNßcat mouse mammary epithelium was assessed with 3D-culture and transplantation assays. RESULTS: Histological and microarray analyses of the mammary lesions of K5ΔNßcat females revealed their high similarity to a subset of basal-like human breast tumors with squamous differentiation. As in human basal-like carcinomas, the Myc pathway appeared to be activated in the mammary lesions of K5ΔNßcat mice. We found that a basal cell population with stem/progenitor characteristics was amplified in K5ΔNßcat mouse preneoplastic glands. Finally, the deletion of Myc from the mammary basal layer of K5ΔNßcat mice not only abolished the regenerative capacity of basal epithelial cells, but, in addition, completely prevented the tumorigenesis. CONCLUSIONS: These results strongly indicate that ß-catenin-induced stem cell amplification and tumorigenesis rely ultimately on the Myc pathway activation and reinforce the hypothesis that basal stem/progenitor cells may be at the origin of a subset of basal-like breast tumors.


Asunto(s)
Neoplasias Mamarias Experimentales/metabolismo , Células Madre Neoplásicas/fisiología , Proteínas Proto-Oncogénicas c-myc/metabolismo , beta Catenina/metabolismo , Animales , Carcinogénesis/metabolismo , Células Epiteliales/metabolismo , Femenino , Humanos , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Trasplante de Neoplasias , Proteínas Proto-Oncogénicas c-myc/genética , Eliminación de Secuencia , Células Tumorales Cultivadas , Vía de Señalización Wnt , beta Catenina/genética
18.
Stem Cells ; 31(9): 1857-67, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23712598

RESUMEN

Mammary epithelium comprises a layer of luminal cells and a basal myoepithelial cell layer. Both mammary epithelial compartments, basal and luminal, contain stem and progenitor cells, but only basal cells are capable of gland regeneration upon transplantation. Aberrant expansion of stem/progenitor cell populations is considered to contribute to breast tumorigenesis. Germline deletions of p53 in humans and mice confer a predisposition to tumors, and stem cell frequency is abnormally high in the mammary epithelium of p53-deficient mice. However, it is unknown whether stem/progenitor cell amplification occurs in both, basal and luminal cell populations in p53-deficient mammary tissue. We used a conditional gene deletion approach to study the role of p53 in stem/progenitor cells residing in the mammary luminal and basal layers. Using two- and three-dimensional cell culture assays, we showed that p53 loss led to the expansion of clonogenic stem/progenitor cells in both mammary epithelial cell layers. Moreover, following p53 deletion, luminal and basal stem/progenitor cells acquired a capacity for unlimited propagation in mammosphere culture. Furthermore, limiting dilution and serial transplantation assays revealed amplification and enhanced self-renewal in the basal regenerating cell population of p53-deficient mammary epithelium. Our data suggest that the increase in stem/progenitor cell activity may be, at least, partially mediated by the Notch pathway. Taken together, these results strongly indicate that p53 restricts the propagation and self-renewal of stem/progenitor cells in both layers of the mammary epithelium providing further insight into the impact of p53 loss in breast cancerogenesis.


Asunto(s)
Compartimento Celular , Células Epiteliales/citología , Glándulas Mamarias Animales/citología , Células Madre/citología , Células Madre/metabolismo , Proteína p53 Supresora de Tumor/deficiencia , Animales , Recuento de Células , Proliferación Celular , Células Clonales , Células Epiteliales/metabolismo , Femenino , Eliminación de Gen , Humanos , Integrasas/metabolismo , Queratina-5/genética , Ratones , Regiones Promotoras Genéticas/genética , Receptores Notch/metabolismo , Esferoides Celulares/citología , Esferoides Celulares/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
19.
Semin Cell Dev Biol ; 23(5): 599-605, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22430758

RESUMEN

Integrins are ubiquitously expressed major cell surface receptors for extracellular matrix. Integrin interaction with their extracellular ligands triggers activation of the intracellular signaling pathways that control cell shape, motility, proliferation, survival, cell-type-specific gene expression. In this review, we summarize recent studies analyzing contribution of integrins to the control of the mammary morphogenesis and differentiation, function and maintenance of mammary stem and progenitor cells and resume the data from mouse models revealing the contribution of the integrin-mediated signaling to mammary tumorigenesis.


Asunto(s)
Integrinas/metabolismo , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Humanas/metabolismo , Animales , Transformación Celular Neoplásica/metabolismo , Humanos , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Humanas/citología , Glándulas Mamarias Humanas/crecimiento & desarrollo , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo
20.
Stem Cells ; 30(6): 1246-54, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22438054

RESUMEN

The mammary epithelium comprises two major cell lineages: basal and luminal. Basal cells (BCs) isolated from the mammary epithelium and transplanted into the mouse mammary fat pad cleared from the endogenous epithelium regenerate the mammary gland, strongly suggesting that the basal epithelial compartment harbors a long-lived cell population with multipotent stem cell potential. The luminal cell layer is devoid of the regenerative potential, but it contains cells with clonogenic capacity, the luminal progenitors. Mammary BCs and luminal progenitors express high levels of the transcription factor Myc. Here, we show that deletion of Myc from mammary basal epithelial cells led to impaired stem cell self-renewal as evaluated by limiting dilution and serial transplantation assays. Luminal progenitor population was significantly diminished in mutant epithelium suggesting control by the BC layer. Colony formation assay performed with isolated BCs showed that clonogenic capacity was abolished by Myc deletion. Moreover, transplanted BCs depleted of Myc failed to produce epithelial outgrowths. Stimulation with ovarian hormones estrogen (E) and progesterone (P) partially rescued the repopulation capacity of Myc-depleted BCs; however, the Myc-deficient mammary epithelium developed in response to E/P treatment lacked stem and progenitor cells. This study provides the first evidence that in the mammary gland, Myc has an essential nonredundant function in the maintenance of the self-renewing multipotent stem cell population responsible for the regenerative capacity of the mammary epithelium and is required downstream from ovarian hormones, for the control of mammary stem and progenitor cell functions.


Asunto(s)
Glándulas Mamarias Animales/citología , Proteínas Proto-Oncogénicas c-myc/fisiología , Células Madre/fisiología , Animales , Diferenciación Celular , Células Epiteliales/citología , Células Epiteliales/fisiología , Células Epiteliales/trasplante , Femenino , Glándulas Mamarias Animales/fisiología , Ratones , Ratones Transgénicos , Células Madre/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA