Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 185(12): 2164-2183.e25, 2022 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-35597241

RESUMEN

X inactivation (XCI) is triggered by upregulation of XIST, which coats the chromosome in cis, promoting formation of a heterochromatic domain (Xi). XIST role beyond initiation of XCI is only beginning to be elucidated. Here, we demonstrate that XIST loss impairs differentiation of human mammary stem cells (MaSCs) and promotes emergence of highly tumorigenic and metastatic carcinomas. On the Xi, XIST deficiency triggers epigenetic changes and reactivation of genes overlapping Polycomb domains, including Mediator subunit MED14. MED14 overdosage results in increased Mediator levels and hyperactivation of the MaSC enhancer landscape and transcriptional program, making differentiation less favorable. We further demonstrate that loss of XIST and Xi transcriptional instability is common among human breast tumors of poor prognosis. We conclude that XIST is a gatekeeper of human mammary epithelium homeostasis, thus unveiling a paradigm in the control of somatic cell identity with potential consequences for our understanding of gender-specific malignancies.


Asunto(s)
Complejo Mediador/metabolismo , Células Madre Neoplásicas/metabolismo , ARN Largo no Codificante/metabolismo , Neoplasias de la Mama/metabolismo , Diferenciación Celular , Epigénesis Genética , Humanos , ARN Largo no Codificante/genética , Inactivación del Cromosoma X
2.
Genes Dev ; 28(20): 2205-18, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25260709

RESUMEN

Signals from the niche play pivotal roles in regulating adult stem cell self-renewal. Previous studies indicated that the steroid hormones can expand mammary stem cells (MaSCs) in vivo. However, the facilitating local niche factors that directly contribute to the MaSC expansion remain unclear. Here we identify R-spondin1 (Rspo1) as a novel hormonal mediator in the mammary gland. Pregnancy and hormonal treatment up-regulate Rspo1 expression. Rspo1 cooperates with another hormonal mediator, Wnt4, to promote MaSC self-renewal through Wnt/ß-catenin signaling. Knockdown of Rspo1 and Wnt4 simultaneously abolishes the stem cell reconstitution ability. In culture, hormonal treatment that stimulates the expression of both Rspo1 and Wnt4 can completely substitute for exogenous Wnt proteins, potently expand MaSCs, and maintain their full development potential in transplantation. Our data unveil the intriguing concept that hormones induce a collaborative local niche environment for stem cells.


Asunto(s)
Diferenciación Celular , Células Madre/citología , Trombospondinas/metabolismo , Animales , Células Cultivadas , Femenino , Técnicas de Silenciamiento del Gen , Ratones Endogámicos BALB C , Transducción de Señal , Trombospondinas/genética , Regulación hacia Arriba , Proteína Wnt4/genética , Proteína Wnt4/metabolismo
3.
Genes Dev ; 28(11): 1143-58, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24888586

RESUMEN

The mammary epithelium is highly responsive to local and systemic signals, which orchestrate morphogenesis of the ductal tree during puberty and pregnancy. Based on transplantation and lineage tracing studies, a hierarchy of stem and progenitor cells has been shown to exist among the mammary epithelium. Lineage tracing has highlighted the existence of bipotent mammary stem cells (MaSCs) in situ as well as long-lived unipotent cells that drive morphogenesis and homeostasis of the ductal tree. Moreover, there is accumulating evidence for a heterogeneous MaSC compartment comprising fetal MaSCs, slow-cycling cells, and both long-term and short-term repopulating cells. In parallel, diverse luminal progenitor subtypes have been identified in mouse and human mammary tissue. Elucidation of the normal cellular hierarchy is an important step toward understanding the "cells of origin" and molecular perturbations that drive breast cancer.


Asunto(s)
Diferenciación Celular , Glándulas Mamarias Animales/citología , Glándulas Mamarias Humanas/citología , Células Madre/citología , Animales , Linaje de la Célula , Femenino , Humanos , Trasplante de Células Madre
4.
J Mammary Gland Biol Neoplasia ; 25(2): 85-102, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32323111

RESUMEN

Breast cancer initiation and progression are often observed as the result of dysregulation of normal developmental processes and pathways. Studies focused on normal mammary stem/progenitor cell activity have led to an understanding of how breast cancer cells acquire stemness-associated properties including tumor initiation, survival and multi-lineage differentiation into heterogeneous tumors that become difficult to target therapeutically. Importantly, more recent investigations have provided valuable insight into how key developmental regulators can impact multiple phases of metastasis, where they are repurposed to not only promote metastatic phenotypes such as migration, invasion and EMT at the primary site, but also to regulate the survival, initiation and maintenance of metastatic lesions at secondary organs. Herein, we discuss findings that have led to a better understanding of how embryonic and pluripotency factors contribute not only to normal mammary development, but also to metastatic progression. We further examine the therapeutic potential of targeting these developmental pathways, and discuss how a better understanding of compensatory mechanisms, crosstalk between pathways, and novel experimental models could provide critical insight into how we might exploit embryonic and pluripotency regulators to inhibit tumor progression and metastasis.


Asunto(s)
Neoplasias de la Mama/patología , Mama/citología , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Células Madre Pluripotentes/citología , Mama/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Diferenciación Celular , Femenino , Humanos , Metástasis de la Neoplasia , Células Madre Pluripotentes/metabolismo , Transducción de Señal
5.
Int J Cancer ; 144(9): 2239-2253, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30450584

RESUMEN

Mammary gland luminal cells are maintained by the proliferation of ER- luminal progenitor (LP) cells. Human breast LP cells exhibit telomere DNA damage, which is associated with mammographic density and increased cancer risk. Telomeric repeat factor 2 (TRF2) protects telomeres from DNA damage response. TRF2 expression is reduced in human breast cancers. We deleted TRF2 expression in mammary gland epithelium. Mammary glands lacking TRF2 expression exhibited increased telomere DNA damage response, histopathological and functional degeneration, and prominent ductal fibrosis. TRF2-deficient mammary tumors exhibited rapid onset and increased proliferation. Tumor derived LP cells failed to form tumors after transplantation. The MSC population was highly tumorigenic and maintained telomeres via the ALT mechanism. Telomere DNA damage response in mammary tumors resulted in p53 dependent ER+ cellular differentiation and sensitivity to anti-estrogen therapy. Our results provide a new in vivo model of mammographic density, stem cell differentiation, cancer risk, and therapeutic sensitivity.


Asunto(s)
Neoplasias de la Mama/patología , Fibrosis/patología , Glándulas Mamarias Animales/patología , Telómero/fisiología , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Daño del ADN/genética , Reparación del ADN/genética , Femenino , Humanos , Glándulas Mamarias Animales/citología , Ratones , Ratones Transgénicos , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Proteína p53 Supresora de Tumor/metabolismo
6.
J Pathol ; 246(4): 497-507, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30101462

RESUMEN

Krüpple-like factor 5 (KLF5) is required for the development of the embryo and multiple organs, such as the lung and intestine. KLF5 plays a pro-proliferative and oncogenic role in several carcinomas, including breast cancer. However, its role in normal mammary gland development and oncogenesis has not been elucidated in vivo. In this study, we used mammary gland-specific Klf5 conditional knockout mice derived by mating Klf5-LoxP and MMTV-Cre mice. The genetic ablation of Klf5 suppresses mammary gland ductal elongation and lobuloalveolar formation. Klf5 deficiency inhibits mammary epithelial cell proliferation, survival, and stem cell maintenance. Klf5 promotes mammary stemness, at least partially, by directly promoting the transcription of Slug. Finally, Klf5 depletion suppressed PyMT-induced mammary gland tumor cell stemness, tumor initiation, and growth in vivo. Slug also mediated these functions of Klf5 in vivo. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/metabolismo , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Células Madre Neoplásicas/metabolismo , Animales , Línea Celular , Proliferación Celular , Femenino , Humanos , Factores de Transcripción de Tipo Kruppel/deficiencia , Factores de Transcripción de Tipo Kruppel/genética , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Morfogénesis , Células Madre Neoplásicas/patología , Fenotipo , Transducción de Señal , Factores de Transcripción de la Familia Snail/genética , Factores de Transcripción de la Familia Snail/metabolismo , Carga Tumoral
7.
J Pathol ; 244(1): 61-70, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28940516

RESUMEN

It is widely accepted that the cell of origin of breast cancer is the adult mammary epithelial stem cell; however, demonstrating the presence and location of tissue stem cells in the human breast has proved difficult. Furthermore, we do not know the clonal architecture of the normal and premalignant mammary epithelium or its cellular hierarchy. Here, we use deficiency in the mitochondrial enzyme cytochrome c oxidase (CCO), typically caused by somatic mutations in the mitochondrial genome, as a means to perform lineage tracing in the human mammary epithelium. PCR sequencing of laser-capture microdissected cells in combination with immunohistochemistry for markers of lineage differentiation was performed to determine the clonal nature of the mammary epithelium. We have shown that in the normal human breast, clonal expansions (defined here by areas of CCO deficiency) are typically uncommon and of limited size, but can occur at any site within the adult mammary epithelium. The presence of a stem cell population was shown by demonstrating multi-lineage differentiation within CCO-deficient areas. Interestingly, we observed infrequent CCO deficiency that was restricted to luminal cells, suggesting that niche succession, and by inference stem cell location, is located within the luminal layer. CCO-deficient areas appeared large within areas of ductal carcinoma in situ, suggesting that the rate of clonal expansion was altered in the premalignant lesion. © 2017 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Asunto(s)
Neoplasias de la Mama/patología , Carcinoma Intraductal no Infiltrante/patología , Linaje de la Célula , Células Madre/fisiología , Mama/patología , Diferenciación Celular , Células Clonales , Complejo IV de Transporte de Electrones/genética , Células Epiteliales/fisiología , Epitelio/patología , Femenino , Humanos , Mitocondrias/enzimología , Lesiones Precancerosas
8.
Biochem Cell Biol ; 96(5): 539-547, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29620414

RESUMEN

Stem cell biology offers promise for understanding the origins of the mammary gland. However, the distribution of mammary stem cell (MaSC) activities at earlier embryonic stages has not been fully identified. The markers for sorting adult MaSC, CD24, CD29, and CD49f have been applied to analyze fetal MaSCs. Here we explored mammary anlagen MaSCs by investigating the expression of CD24 and CD49f. According to the comparative analysis between adult mammary gland and fetal mammary anlagen, we found that fetal mouse mammary anlagen may possess a high percentage of potential MaSCs. Flow cytometry analysis revealed 2 distinct mammary anlagen populations: Lin-CD24med and Lin-CD24high. Sphere-forming and mammary repopulating assays confirmed that the stem cell activity of E14.5 mouse mammary anlagen was restricted to the Lin-CD24med cell population. Furthermore, CD24med mammary anlagen cells were separated into Lin-CD24medCD49f+ and Lin-CD24medCD49f- populations and identified, respectively. The results proved that the mammary anlagen Lin-CD24medCD49f+ cell population possesses more stem cell activities than the Lin-CD24medCD49f- cell population. However, a limited numbers of stem cells and large numbers of stromal cells were identified in mammary anlagen in the Lin-CD24med cell population.


Asunto(s)
Antígeno CD24/metabolismo , Embrión de Mamíferos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Integrina alfa6/metabolismo , Glándulas Mamarias Animales/embriología , Células Madre Embrionarias de Ratones/metabolismo , Animales , Embrión de Mamíferos/citología , Femenino , Glándulas Mamarias Animales/citología , Ratones , Células Madre Embrionarias de Ratones/citología
9.
Development ; 142(18): 3239-51, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26071498

RESUMEN

We present an organoid regeneration assay in which freshly isolated human mammary epithelial cells are cultured in adherent or floating collagen gels, corresponding to a rigid or compliant matrix environment. In both conditions, luminal progenitors form spheres, whereas basal cells generate branched ductal structures. In compliant but not rigid collagen gels, branching ducts form alveoli at their tips, express basal and luminal markers at correct positions, and display contractility, which is required for alveologenesis. Thereby, branched structures generated in compliant collagen gels resemble terminal ductal-lobular units (TDLUs), the functional units of the mammary gland. Using the membrane metallo-endopeptidase CD10 as a surface marker enriches for TDLU formation and reveals the presence of stromal cells within the CD49f(hi)/EpCAM(-) population. In summary, we describe a defined in vitro assay system to quantify cells with regenerative potential and systematically investigate their interaction with the physical environment at distinct steps of morphogenesis.


Asunto(s)
Biomarcadores/metabolismo , Técnicas de Cultivo de Célula/métodos , Glándulas Mamarias Humanas/citología , Glándulas Mamarias Humanas/fisiología , Morfogénesis/fisiología , Organoides/fisiología , Regeneración/fisiología , Separación Celular/métodos , Colágeno , Femenino , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Humanos , Técnicas de Dilución del Indicador , Neprilisina/metabolismo
10.
Cancer Metastasis Rev ; 35(4): 631-644, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27866304

RESUMEN

The mammary epithelium is organized in a hierarchy of mammary stem cells (MaSCs), progenitors, and differentiated cells. The development and homeostasis of mammary gland are tightly controlled by a complex network of cell lineage regulators. These determinants of cellular hierarchy are frequently deregulated in breast tumor cells and closely associated with cancer progression and metastasis. They also contribute to the diversity of breast cancer subtypes and their distinct metastatic patterns. Cell fate regulators that normally promote stem/progenitor activities can serve as drivers for epithelial-mesenchymal transition and metastasis whereas regulators that promote terminal differentiation generally suppress metastasis. In this review, we discuss how some of the key factors function in normal mammary lineage determination and how these processes are hijacked by tumor cells to enhance metastasis. Understanding the molecular connections between normal development and cancer metastasis will enable the development of more specific and effective therapeutic approaches targeting metastatic tumor cells.


Asunto(s)
Neoplasias de la Mama/patología , Animales , Linaje de la Célula , Femenino , Humanos , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/patología , Glándulas Mamarias Humanas/citología , Glándulas Mamarias Humanas/patología , Metástasis de la Neoplasia
11.
Cell Biol Int ; 40(11): 1212-1223, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27590622

RESUMEN

Women with high mammographic density (MD) are at increased risk of breast cancer (BC) after adjustment for age and body mass index. We have developed a murine biochamber model in which both high MD (HMD) and low MD (LMD) tissue can be propagated. Here, we tested whether cells isolated by collagenase digestion and fluorescence-activated cell sorting (FACS) from normal breast can be reconstituted in our biochamber model, which would allow cell-specific manipulations to be tested. Fresh breast tissue was collected from women (n = 7) undergoing prophylactic mastectomy. The tissue underwent collagenase digestion overnight and, in some cases, additional FACS enrichment to obtain mature epithelial, luminal progenitor, mammary stem, and stromal cells. Cells were then transferred bilaterally into biochambers in SCID mice (n = 5-7) and incubated for 6 weeks, before harvesting for histological analyses, and immunohistochemical staining for cytokeratins (CK), vimentin, Ki-67, murine macrophages, and Cleaved Caspase-3. Biochambers inoculated with single cells after collagenase digestion or with flow cytometry contained glandular structures of human origin (human vimentin-positive), which expressed CK-14 and pan-CK, and were proliferating (Ki-67-positive). Glandular structures from the digested tissues were smaller than those in chambers seeded with finely chopped intact mammary tissue. Mouse macrophage infiltration was higher in the chambers arising from digested tissues. Pooled single cells and FACS fractionated cells were viable in the murine biochambers and formed proliferating glandular organoids of human origin. This is among the first report to demonstrate the success of formed human glandular organoids from isolated primary mammary cells in the murine biochamber model.


Asunto(s)
Mama/crecimiento & desarrollo , Colagenasas/metabolismo , Organoides/crecimiento & desarrollo , Ingeniería de Tejidos/métodos , Adulto , Animales , Mama/citología , Mama/metabolismo , Densidad de la Mama , Neoplasias de la Mama/patología , Proliferación Celular/fisiología , Colagenasas/química , Femenino , Citometría de Flujo/métodos , Humanos , Glándulas Mamarias Humanas/citología , Glándulas Mamarias Humanas/crecimiento & desarrollo , Ratones , Ratones SCID , Persona de Mediana Edad , Organoides/citología , Organoides/metabolismo , Cultivo Primario de Células
12.
J Biol Chem ; 289(45): 31310-8, 2014 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-25231996

RESUMEN

Long noncoding RNAs (lncRNAs) have emerged as key regulators of gene expression in embryonic stem cell (ESC) self-renewal and differentiation. In ESCs, lncRNAs are regulated at the genetic level via transcription factor binding to lncRNA gene promoters. Here we demonstrate that the key cytoprotective transcription factor NRF2 controls lncRNA expression in mammary stem cells. By profiling lncRNAs in wild-type and NRF2 knockdown mammary stem cells, we demonstrate that the lncRNA ROR, a regulator of embryonic stem cell pluripotency, is overexpressed upon NRF2 knockdown. We performed promoter analyses and examined predicted NRF2 binding elements in the ROR promoter using luciferase reporter constructs of a ROR promoter deletion series. Our studies revealed that NRF2 binds to two specific NRF2 response elements flanking the ROR promoter and that these two NRF2 response elements are equally important to suppress ROR transcription. In addition, we identified associated H3K27me3 chromatin modification and EZH2 binding at the ROR promoter that was dependent on NRF2 binding. We observed that NRF2 knockdown or ROR overexpression leads to increased stem cell self-renewal in mammary stem cells. Furthermore, we demonstrate Nrf2 regulation of the mammary stem cell population in vivo. These observations provide further evidence for the critical role of NRF2 in maintaining normal stem cell subpopulations in mammary epithelium.


Asunto(s)
Estrógenos/metabolismo , Regulación Neoplásica de la Expresión Génica , Factor 2 Relacionado con NF-E2/metabolismo , ARN Largo no Codificante , Células Madre/citología , Animales , Antioxidantes/química , Línea Celular Tumoral , Células Madre Embrionarias/citología , Células Epiteliales/citología , Femenino , Hematopoyesis , Humanos , Hibridación Fluorescente in Situ , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Regiones Promotoras Genéticas , Unión Proteica , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Transducción de Señal
13.
Stem Cells ; 32(3): 649-61, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24038768

RESUMEN

Children exposed to ionizing radiation have a substantially greater breast cancer risk than adults; the mechanism for this strong age dependence is not known. Here we show that pubertal murine mammary glands exposed to sparsely or densely ionizing radiation exhibit enrichment of mammary stem cell and Notch pathways, increased mammary repopulating activity indicative of more stem cells, and propensity to develop estrogen receptor (ER) negative tumors thought to arise from stem cells. We developed a mammary lineage agent-based model (ABM) to evaluate cell inactivation, self-renewal, or dedifferentiation via epithelial-mesenchymal transition (EMT) as mechanisms by which radiation could increase stem cells. ABM rejected cell inactivation and predicted increased self-renewal would only affect juveniles while dedifferentiation could act in both juveniles and adults. To further test self-renewal versus dedifferentiation, we used the MCF10A human mammary epithelial cell line, which recapitulates ductal morphogenesis in humanized fat pads, undergoes EMT in response to radiation and transforming growth factor ß (TGFß) and contains rare stem-like cells that are Let-7c negative or express both basal and luminal cytokeratins. ABM simulation of population dynamics of double cytokeratin cells supported increased self-renewal in irradiated MCF10A treated with TGFß. Radiation-induced Notch concomitant with TGFß was necessary for increased self-renewal of Let-7c negative MCF10A cells but not for EMT, indicating that these are independent processes. Consistent with these data, irradiating adult mice did not increase mammary repopulating activity or ER-negative tumors. These studies suggest that irradiation during puberty transiently increases stem cell self-renewal, which increases susceptibility to developing ER-negative breast cancer.


Asunto(s)
Envejecimiento/patología , Glándulas Mamarias Animales/patología , Glándulas Mamarias Animales/efectos de la radiación , Neoplasias Mamarias Animales/patología , Radiación Ionizante , Receptores de Estrógenos/metabolismo , Células Madre/patología , Animales , Biomarcadores/metabolismo , Línea Celular , Linaje de la Célula , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Simulación por Computador , Relación Dosis-Respuesta en la Radiación , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Células Epiteliales/efectos de la radiación , Femenino , Humanos , Neoplasias Mamarias Animales/metabolismo , Ratones , Morfogénesis/efectos de los fármacos , Morfogénesis/efectos de la radiación , Receptores Notch/metabolismo , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Células Madre/efectos de la radiación , Factor de Crecimiento Transformador beta/farmacología
14.
Cell Biol Int ; 39(5): 611-8, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25572907

RESUMEN

Fresh human breast milk consists of a heterogeneous population of cells that may offer a non-invasive source of cells for therapeutic proposes. The aims of this study were to characterize the breast milk-derived cells cultured in vitro. To do this, the cells from human breast milk were cultured and the expression of the CD markers along with the embryonic stem cell markers, endothelial and luminal mammary epithelial cell markers was evaluated by flow cytometry and immunofluorescence. The presence of fetal microchimerism among the isolated cells was also determined by the presence of SRY gene. They were also differentiated into adipocytes and osteoblasts. The results showed that a remarkable number of cells expressed the mesenchymal stem cell (MSC) markers such as CD90, CD44, CD271, and CD146. A subpopulation of the human breast milk-derived cells (HBMDC) also expressed the embryonic stem cell markers, such as TRA 60-1, Oct4, Nanog and Sox2 but not SSEA1 or 4. The frequencies of the cells which expressed the endothelial, hematopoietic cell markers were negligible. SRY gene was not detected in the breast milk isolated cells. A subpopulation of the cells also expressed cytokeratin 18, the marker of luminal mammary epithelial cells. These cells showed the capability to differentiate into adipocytes and osteoblasts. In conclusion, these finding highlighted the presence of cells with various sources in the breast milk. Different stem cells including MSCs or embryonic stem cell-like cell along with the exfoliated cells from luminal epithelial cells were found among the isolated cells. The breast milk-derived stem cells might be considered as a non-invasive source of the stem cells for therapeutic purpose.


Asunto(s)
Células Madre Mesenquimatosas/citología , Leche Humana/citología , Adipocitos/fisiología , Adulto , Diferenciación Celular , Separación Celular/métodos , Células Cultivadas , Células Epiteliales/citología , Células Epiteliales/fisiología , Femenino , Citometría de Flujo , Humanos , Glándulas Mamarias Humanas/citología , Células Madre Mesenquimatosas/fisiología , Osteoblastos/fisiología , Adulto Joven
15.
Stem Cells ; 31(9): 1954-65, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23766243

RESUMEN

Receptor Activator of NF-kappa B (RANK) pathway controls mammary gland development in mice but its role in mammary stem cell fate remains undefined. We show that constitutive RANK signaling expands luminal and basal mammary compartments including mammary stem and luminal progenitor cell pools and interferes with the generation of CD61+ and Sca1+ luminal cells and Elf5 expression. Impaired mammary cell commitment upon RANK overexpression leads to the accumulation of progenitors including K14+K8+ bipotent cells and the formation of heterogeneous tumors containing hyperplastic basal, luminal, and progenitor cells. RANK expression increases in wild-type mammary epithelia with age and parity, and spontaneous preneoplastic lesions express RANK and accumulate K14+K8+ cells. In human breast tumors, high RANK expression levels are also associated with altered mammary differentiation. These results suggest that increased RANK signaling interferes with mammary cell commitment, contributing to breast carcinogenesis.


Asunto(s)
Carcinogénesis/patología , Linaje de la Célula , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patología , Envejecimiento/patología , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinogénesis/genética , Compartimento Celular , Diferenciación Celular , Forma de la Célula , Epitelio/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Queratinas/metabolismo , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Virus del Tumor Mamario del Ratón/fisiología , Ratones , Modelos Biológicos , Paridad , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Embarazo , Receptor Activador del Factor Nuclear kappa-B/genética , Células Madre/metabolismo
16.
Stem Cells ; 31(9): 1921-31, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23712846

RESUMEN

The key signaling networks regulating mammary stem cells are poorly defined. The leucine-rich repeat containing G protein-coupled receptor (Lgr) family has been implicated in intestinal, gastric, and epidermal stem cell functions. We investigated whether Lgr4 functions in mammary gland development and mammary stem cells. We found that Lgr4(-/-) mice had delayed ductal development, fewer terminal end buds, and decreased side-branching. Crucially, the mammary stem cell repopulation capacity was severely impaired. Mammospheres from Lgr4(-/-) mice showed decreased Wnt signaling. Wnt3a treatment prevented the adverse effects of Lgr4 loss on organoid formation. Chromatin immunoprecipitation analysis indicated that Sox2 expression was controlled by the Lgr4/Wnt/ß-catenin/Lef1 pathway. Importantly, Sox2 overexpression restored the in vivo mammary regeneration potential of Lgr4(-/-) mammary stem cells. Therefore, Lgr4 activates Sox2 to regulate mammary development and stem cell functions via Wnt/ß-catenin/Lef1.


Asunto(s)
Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/crecimiento & desarrollo , Células Madre Pluripotentes/citología , Receptores Acoplados a Proteínas G/metabolismo , Factores de Transcripción SOXB1/metabolismo , Animales , Linaje de la Célula , Proliferación Celular , Ensayo de Unidades Formadoras de Colonias , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Humanos , Glándulas Mamarias Animales/metabolismo , Ratones , Ratones Endogámicos C57BL , Morfogénesis , Células Madre Multipotentes/citología , Organoides/citología , Organoides/metabolismo , Fenotipo , Células Madre Pluripotentes/metabolismo , Receptores Acoplados a Proteínas G/deficiencia , Regeneración , Vía de Señalización Wnt
17.
Stem Cells Dev ; 33(17-18): 449-467, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38943275

RESUMEN

The origin of breast cancer (BC) has traditionally been a focus of medical research. It is widely acknowledged that BC originates from immortal mammary stem cells and that these stem cells participate in two division modes: symmetric cell division (SCD) and asymmetrical cell division (ACD). Although both of these modes are key to the process of breast development and their imbalance is closely associated with the onset of BC, the molecular mechanisms underlying these phenomena deserve in-depth exploration. In this review, we first outline the molecular mechanisms governing ACD/SCD and analyze the role of ACD/SCD in various stages of breast development. We describe that the changes in telomerase activity, the role of polar proteins, and the stimulation of ovarian hormones subsequently lead to two distinct consequences: breast development or carcinogenesis. Finally, gene mutations, abnormalities in polar proteins, modulation of signal-transduction pathways, and alterations in the microenvironment disrupt the balance of BC stem cell division modes and cause BC. Important regulatory factors such as mammalian Inscuteable mInsc, Numb, Eya1, PKCα, PKCθ, p53, and IL-6 also play significant roles in regulating pathways of ACD/SCD and may constitute key targets for future research on stem cell division, breast development, and tumor therapy.


Asunto(s)
Neoplasias de la Mama , Carcinogénesis , Glándulas Mamarias Humanas , Humanos , Femenino , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/genética , Animales , Glándulas Mamarias Humanas/crecimiento & desarrollo , Glándulas Mamarias Humanas/patología , Glándulas Mamarias Humanas/citología , Glándulas Mamarias Humanas/metabolismo , Carcinogénesis/patología , Carcinogénesis/metabolismo , Carcinogénesis/genética , Células Madre/metabolismo , Células Madre/citología , División Celular , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/patología , Glándulas Mamarias Animales/metabolismo , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Transducción de Señal
18.
Cell Rep ; 43(9): 114762, 2024 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-39321020

RESUMEN

Adult mammary stem cells (aMaSCs) are vital to tissue expansion and remodeling during the process of postnatal mammary development. The protein C receptor (Procr) is one of the well-identified surface markers of multipotent aMaSCs. However, an understanding of the regulatory mechanisms governing Procr's protein stability remains incomplete. In this study, we identified Glycoprotein m6a (Gpm6a) as a critical protein for aMaSC activity modulation by using the Gpm6a knockout mouse model. Interestingly, we determined that Gpm6a depletion results in a reduction of Procr protein stability. Mechanistically, Gpm6a regulates Procr protein stability by mediating the formation of lipid rafts, a process requiring Zdhhc1 and Zdhhc2 to palmitate Gpm6a at Cys17,18 and Cys246 sites. Our findings highlight an important mechanism involving Zdhhc1- and Zdhhc2-mediated Gpm6a palmitoylation for the regulation of Procr stability, aMaSC activity, and postnatal mammary development.


Asunto(s)
Aciltransferasas , Lipoilación , Glándulas Mamarias Animales , Animales , Aciltransferasas/metabolismo , Aciltransferasas/genética , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/citología , Ratones , Femenino , Ratones Noqueados , Humanos , Microdominios de Membrana/metabolismo , Células Madre/metabolismo , Células Madre/citología , Estabilidad Proteica
19.
Life Sci ; 326: 121781, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37207809

RESUMEN

INTRODUCTION: The cellular behavior in normal mammary gland development and the progression of breast cancer is like the relationship between an object and its mirror image: they may appear similar, but their essence is completely different. Breast cancer can be considered as temporal and spatial aberrations of normal development in mammary gland. Glycans have been shown to regulate key pathophysiological steps during mammary development and breast cancer progression, and the glycoproteins that play a key role in both processes can affect the normal differentiation and development of mammary cells, and even cause malignant transformation or accelerate tumorigenesis due to differences in their type and level of glycosylation. KEY FINDINGS: In this review, we summarize the roles of glycan alterations in essential cellular behaviors during breast cancer progression and mammary development, and also highlight the importance of key glycan-binding proteins such as epidermal growth factor receptor, transforming growth factor ß receptors and other proteins, which are pivotal in the modulation of cellular signaling in mammary gland. Our review takes an overall view of the molecular interplay, signal transduction and cellular behaviors in mammary gland development and breast cancer progression from a glycobiological perspective. SIGNIFICANCE: This review will give a better understanding of the similarities and differences in glycosylation between mammary gland development and breast cancer progression, laying the foundation for elucidating the key molecular mechanisms of glycobiology underlying the malignant transformation of mammary cells.


Asunto(s)
Glándulas Mamarias Humanas , Neoplasias , Animales , Humanos , Glicosilación , Neoplasias/metabolismo , Transformación Celular Neoplásica/metabolismo , Diferenciación Celular , Polisacáridos , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/patología
20.
Cell Rep ; 40(11): 111331, 2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-36103813

RESUMEN

The Wnt/ß-catenin signaling pathway plays an important role in regulating mammary organogenesis and oncogenesis. However, therapeutic methods targeting the Wnt pathway against breast cancer have been limited. To address this challenge, we investigate the function of cyclin-dependent kinase 14 (CDK14), a member of the Wnt signaling pathway, in mammary development and breast cancer progression. We show that CDK14 is expressed in the mammary basal layer and elevated in triple negative breast cancer (TNBC). CDK14 knockdown reduces the colony-formation ability and regeneration capacity of mammary basal cells and inhibits the progression of murine MMTV-Wnt-1 basal-like mammary tumor. CDK14 knockdown or pharmacological inhibition by FMF-04-159-2 suppresses the progression and metastasis of TNBC. Mechanistically, CDK14 inhibition inhibits mammary regeneration and TNBC progression by attenuating Wnt/ß-catenin signaling. These findings highlight the significance of CDK14 in mammary development and TNBC progression, shedding light on CDK14 as a promising therapeutic target for TNBC.


Asunto(s)
Proteínas Quinasas/metabolismo , Neoplasias de la Mama Triple Negativas , Animales , Mama/metabolismo , Línea Celular Tumoral , Quinasas Ciclina-Dependientes/metabolismo , Humanos , Ratones , Células Madre/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Vía de Señalización Wnt
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA