Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
iScience ; 23(11): 101649, 2020 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-33103086

RESUMEN

The receptor tyrosine kinase AXL is associated with epithelial plasticity in several solid tumors including breast cancer and AXL-targeting agents are currently in clinical trials. We hypothesized that AXL is a driver of stemness traits in cancer by co-option of a regulatory function normally reserved for stem cells. AXL-expressing cells in human mammary epithelial ducts co-expressed markers associated with multipotency, and AXL inhibition abolished colony formation and self-maintenance activities while promoting terminal differentiation in vitro. Axl-null mice did not exhibit a strong developmental phenotype, but enrichment of Axl + cells was required for mouse mammary gland reconstitution upon transplantation, and Axl-null mice had reduced incidence of Wnt1-driven mammary tumors. An AXL-dependent gene signature is a feature of transcriptomes in basal breast cancers and reduced patient survival irrespective of subtype. Our interpretation is that AXL regulates access to epithelial plasticity programs in MaSCs and, when co-opted, maintains acquired stemness in breast cancer cells.

2.
Sci Rep ; 10(1): 4788, 2020 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-32161318

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

3.
Sci Rep ; 9(1): 14843, 2019 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-31619692

RESUMEN

Human breast cancer is believed to arise in luminal progenitors within the normal breast. A subset of these are double positive (DP) for basal and luminal keratins and localizes to a putative stem cell zone within ducts. We here present a new protocol based on a combination of CD146 with CD117 and CD326 which provides an up to thirty fold enrichment of the DP cells. We show by expression profiling, colony formation, and morphogenesis that CD146high/CD117high/CD326high DP cells belong to a luminal progenitor compartment. While these DP cells are located quite uniformly in ducts, with age a variant type of DP (vDP) cells, which is mainly CD146-negative, accumulates in lobules. Intriguingly, in specimens with BRCA1 mutations known to predispose for cancer, higher frequencies of lobular vDP cells are observed. We propose that vDP cells are strong candidates for tracing the cellular origin of breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Carcinogénesis , Queratina-14/metabolismo , Queratina-19/metabolismo , Glándulas Mamarias Humanas/metabolismo , Células Madre Neoplásicas/metabolismo , Adolescente , Adulto , Neoplasias de la Mama/patología , Antígeno CD146/metabolismo , Células Cultivadas , Femenino , Voluntarios Sanos , Humanos , Glándulas Mamarias Humanas/patología , Persona de Mediana Edad , Células Madre Neoplásicas/patología , Adulto Joven
4.
Am J Pathol ; 184(4): 1198-208, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24655379

RESUMEN

Elucidating the phenotypic evolution of breast cancer through distinct subtypes relies heavily on defining a lineage blueprint of the normal human breast. Here, we show that in normal breast, within the luminal epithelial lineage, a subset of cells characterized by strong staining for endocrine receptors are also characterized by expression of the surface marker CEACAM6. Topographically, this pattern of staining predominates in terminal ductal lobular units, rather than in interlobular ducts. In culture, CEACAM6-expressing cells remain essentially postmitotic under conditions in which the other cells of luminal epithelial lineage are highly proliferative. We examined the pattern of expression among three major breast cancer subtypes: luminal, HER2-enriched, and basal-like. In 104 biopsies, the luminal and HER2-enriched subtypes showed a high proportion of CEACAM6(+) tumors (78% and 83%, respectively); the basal-like subtype showed a low proportion (28%). Further accentuation of this pattern was observed in 13 established breast cancer cell lines. When differentiation was induced by all-trans retinoic acid, CEACAM6 expression strongly correlated with luminal-like differentiation. Furthermore, CEACAM6(+) cancer cells were less proliferative than CEACAM6(-) cells in tumorsphere assays and were less tumorigenic in nude mice. Based on these observations, we propose that luminal and HER2-enriched breast cancers are more closely related than previously thought and may share a common cell of origin.


Asunto(s)
Antígenos CD/biosíntesis , Biomarcadores de Tumor/análisis , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Moléculas de Adhesión Celular/biosíntesis , Receptor ErbB-2/metabolismo , Animales , Western Blotting , Femenino , Citometría de Flujo , Proteínas Ligadas a GPI/biosíntesis , Xenoinjertos , Humanos , Inmunohistoquímica , Ratones , Ratones Desnudos , Reacción en Cadena en Tiempo Real de la Polimerasa
5.
PLoS One ; 7(9): e46543, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23029547

RESUMEN

INTRODUCTION: Solid tumors are less oxygenated than their tissue of origin. Low intra-tumor oxygen levels are associated with worse outcome, increased metastatic potential and immature phenotype in breast cancer. We have reported that tumor hypoxia correlates to low differentiation status in breast cancer. Less is known about effects of hypoxia on non-malignant cells. Here we address whether hypoxia influences the differentiation stage of non-malignant breast epithelial cells and potentially have bearing on early stages of tumorigenesis. METHODS: Normal human primary breast epithelial cells and immortalized non-malignant mammary epithelial MCF-10A cells were grown in a three-dimensional overlay culture on laminin-rich extracellular matrix for up to 21 days at normoxic or hypoxic conditions. Acinar morphogenesis and expression of markers of epithelial differentiation and cell polarization were analyzed by immunofluorescence, immunohistochemistry, qPCR and immunoblot. RESULTS: In large ductal carcinoma in situ patient-specimens, we find that epithelial cells with high HIF-1α levels and multiple cell layers away from the vasculature are immature compared to well-oxygenated cells. We show that hypoxic conditions impaired acinar morphogenesis of primary and immortalized breast epithelial cells grown ex vivo on laminin-rich matrix. Normoxic cultures formed polarized acini-like spheres with the anticipated distribution of marker proteins associated with mammary epithelial polarization e.g. α6-integrin, laminin 5 and Human Milk Fat Globule/MUC1. At hypoxia, cells were not polarized and the sub-cellular distribution pattern of the marker proteins rather resembled that reported in vivo in breast cancer. The hypoxic cells remained in a mitotic state, whereas proliferation ceased with acinar morphogenesis at normoxia. We found induced expression of the differentiation repressor ID1 in the undifferentiated hypoxic MCF-10A cell structures. Acinar morphogenesis was associated with global histone deacetylation whereas the hypoxic breast epithelial cells showed sustained global histone acetylation, which is generally associated with active transcription and an undifferentiated proliferative state.


Asunto(s)
Neoplasias de la Mama/patología , Carcinoma Intraductal no Infiltrante/patología , Transformación Celular Neoplásica/metabolismo , Células Epiteliales/metabolismo , Glándulas Mamarias Humanas/patología , Acetilación , Células Acinares/patología , Antígenos de Diferenciación/metabolismo , Neoplasias de la Mama/metabolismo , Carcinoma Intraductal no Infiltrante/metabolismo , Moléculas de Adhesión Celular/metabolismo , Muerte Celular , Hipoxia de la Célula , Línea Celular , Polaridad Celular , Proliferación Celular , Células Epiteliales/patología , Células Epiteliales/fisiología , Transición Epitelial-Mesenquimal , Matriz Extracelular/fisiología , Femenino , Expresión Génica , Regulación de la Expresión Génica , Glucolípidos/metabolismo , Glicoproteínas/metabolismo , Histonas/metabolismo , Humanos , Proteína 1 Inhibidora de la Diferenciación/genética , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Integrina alfa6/metabolismo , Gotas Lipídicas , Fenotipo , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Kalinina
6.
Mol Med ; 18: 1109-21, 2012 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-22692575

RESUMEN

The CD44(hi) compartment in human breast cancer is enriched in tumor-initiating cells; however, the functional heterogeneity within this subpopulation remains poorly defined. We used a triple-negative breast cancer cell line with a known bilineage phenotype to isolate and clone CD44(hi) single cells that exhibited mesenchymal/basal B and luminal/basal A features, respectively. Herein, we demonstrate in this and other triple-negative breast cancer cell lines that, rather than CD44(hi)/CD24(-) mesenchymal-like basal B cells, the CD44(hi)/CD24(lo) epithelioid basal A cells retained classic cancer stem cell features, such as tumor-initiating capacity in vivo, mammosphere formation and resistance to standard chemotherapy. These results complement previous findings using oncogene-transformed normal mammary cells showing that only cell clones with a mesenchymal phenotype exhibit cancer stem cell features. Further, we performed comparative quantitative proteomic and gene array analyses of these cells and identified potential novel markers of breast cancer cells with tumor-initiating features, such as lipolysis-stimulated lipoprotein receptor (LSR), RAB25, S100A14 and mucin 1 (MUC1), as well as a novel 31-gene signature capable of predicting distant metastasis in cohorts of estrogen receptor-negative human breast cancers. These findings strongly favor functional heterogeneity in the breast cancer cell compartment and hold promise for further refinements of prognostic marker profiling. Our work confirms that, in addition to cancer stem cells with mesenchymal-like morphology, those tumor-initiating cells with epithelial-like morphology should also be the focus of drug development.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Compartimento Celular/genética , Perfilación de la Expresión Génica , Heterogeneidad Genética , Receptores de Hialuranos/metabolismo , Células Madre Neoplásicas/patología , Animales , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Antígeno CD24/metabolismo , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Células Clonales , Resistencia a Antineoplásicos/genética , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Mesodermo/metabolismo , Mesodermo/patología , Ratones , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Fenotipo , Pronóstico , Proteómica , Transcriptoma/genética
7.
Cancer Res ; 72(14): 3687-701, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22552289

RESUMEN

Women older than 50 years account for 75% of new breast cancer diagnoses, and the majority of these tumors are of a luminal subtype. Although age-associated changes, including endocrine profiles and alterations within the breast microenvironment, increase cancer risk, an understanding of the cellular and molecular mechanisms that underlies these observations is lacking. In this study, we generated a large collection of normal human mammary epithelial cell strains from women ages 16 to 91 years, derived from primary tissues, to investigate the molecular changes that occur in aging breast cells. We found that in finite lifespan cultured and uncultured epithelial cells, aging is associated with a reduction of myoepithelial cells and an increase in luminal cells that express keratin 14 and integrin-α6, a phenotype that is usually expressed exclusively in myoepithelial cells in women younger than 30 years. Changes to the luminal lineage resulted from age-dependent expansion of defective multipotent progenitors that gave rise to incompletely differentiated luminal or myoepithelial cells. The aging process therefore results in both a shift in the balance of luminal/myoepithelial lineages and to changes in the functional spectrum of multipotent progenitors, which together increase the potential for malignant transformation. Together, our findings provide a cellular basis to explain the observed vulnerability to breast cancer that increases with age.


Asunto(s)
Envejecimiento , Diferenciación Celular , Senescencia Celular , Glándulas Mamarias Humanas/citología , Células Madre Multipotentes/fisiología , Anciano , Anciano de 80 o más Años , Células Cultivadas , Células Epiteliales/citología , Femenino , Humanos , Persona de Mediana Edad , Fenotipo
8.
Int J Dev Biol ; 55(7-9): 719-29, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22161829

RESUMEN

Distinct subsets of cells, including cells with stem cell-like properties, have been proposed to exist in normal human breast epithelium and breast carcinomas. The cellular origins of epithelial cells contributing to gland development, tissue homeostasis and cancer are, however, still poorly understood. The mouse is a widely used model of mammary gland development, both directly by studying the mouse mammary epithelial cells themselves and indirectly, by studying development, morphogenesis, differentiation and carcinogenesis of xenotransplanted human breast epithelium in vivo. While in early studies, human or mouse epithelium was implanted as fragments into the mouse gland, more recent technical progress has allowed the self-renewal capacity and differentiation potential of distinct cell populations or even individual cells to be interrogated. Here, we review and discuss similarities and differences between mouse and human gland development with particular emphasis on the identity and localization of stem cells, and the influence of the surrounding microenvironment. It is concluded that while recent advances in the field have contributed immense insight into how the normal mammary gland develops and is maintained, significant discrepancies exist between the mouse and human gland which should be taken into consideration in current and future models of mammary stem cell biology.


Asunto(s)
Mama/citología , Glándulas Mamarias Animales/citología , Células Madre/citología , Animales , Mama/crecimiento & desarrollo , Neoplasias de la Mama/patología , Diferenciación Celular , Linaje de la Célula , Femenino , Humanos , Glándulas Mamarias Animales/crecimiento & desarrollo , Ratones , Células Madre Neoplásicas/patología , Transducción de Señal , Nicho de Células Madre , Trasplante de Células Madre , Células Madre/fisiología , Trasplante Heterólogo , Microambiente Tumoral
9.
Ugeskr Laeger ; 172(38): 2600-3, 2010 Sep 20.
Artículo en Danés | MEDLINE | ID: mdl-20920403

RESUMEN

The finding that tumours, like normal tissues, are endowed with varying degrees of cellular heterogeneity has far-reaching consequences for our understanding of cancer. The cancer stem cell and clonal evolution models have both been proposed to explain tumour-associated cellular heterogeneity. Here, we briefly review these two non-exclusive models with special emphasis on how they aid our understanding of cancer and their implications for therapeutic strategies. Finally, we discuss the close association between basic stem cell biology and cancer, focusing on the role of self-renewal.


Asunto(s)
Neoplasias/patología , Células Madre Neoplásicas , Antineoplásicos/uso terapéutico , Descubrimiento de Drogas , Humanos , Modelos Biológicos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Células Madre Neoplásicas/diagnóstico por imagen , Células Madre Neoplásicas/patología , Ultrasonografía
10.
Am J Pathol ; 176(3): 1229-40, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20133812

RESUMEN

Tumor cells can activate stroma, yet the implication of this activation in terms of reciprocal induction of gene expression in tumor cells is poorly understood. Epithelial Stromal Interaction 1 (EPSTI1) is an interferon response gene originally isolated from heterotypic recombinant cultures of human breast cancer cells and activated breast myofibroblasts. Here we describe the first immunolocalization of EPSTI1 in normal and cancerous breast tissue, and we provide evidence for a role of this molecule in the regulation of tumor cell properties and epithelial-mesenchymal transition. In general, no EPSTI1 staining was observed in normal breast epithelial cells from reduction mammoplasties (n=25). However, in carcinomas, staining was positive in 22 of 40 biopsies and inversely correlated with the level of differentiation. To address the function of EPSTI1, we expressed EPSTI1 ectopically in one cell line and silenced endogenous EPSTI1 by RNA interference in another. Irrespective of the experimental approach, EPSTI1 expression led to an increase in tumorsphere formation-a property associated with breast stem/progenitor cells. Most remarkably, we show that EPSTI1, by conveying spread of tumor cells, can replace peritumoral activated fibroblasts in a tumor environment assay. These observations implicate EPSTI1 as a hitherto unappreciated regulator of tumor cell properties.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Proteínas de Neoplasias/metabolismo , Animales , Bioensayo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Epitelio/metabolismo , Epitelio/patología , Femenino , Humanos , Mesodermo/metabolismo , Mesodermo/patología , Ratones , Especificidad de Órganos , Interferencia de ARN
11.
Integr Biol (Camb) ; 1(1): 70-9, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20023793

RESUMEN

In adult tissues, multi-potent progenitor cells are some of the most primitive members of the developmental hierarchies that maintain homeostasis. That progenitors and their more mature progeny share identical genomes, suggests that fate decisions are directed by interactions with extrinsic soluble factors, ECM, and other cells, as well as physical properties of the ECM. To understand regulation of fate decisions, therefore, would require a means of understanding carefully choreographed combinatorial interactions. Here we used microenvironment protein microarrays to functionally identify combinations of cell-extrinsic mammary gland proteins and ECM molecules that imposed specific cell fates on bipotent human mammary progenitor cells. Micropatterned cell culture surfaces were fabricated to distinguish between the instructive effects of cell-cell versus cell-ECM interactions, as well as constellations of signaling molecules; and these were used in conjunction with physiologically relevant 3 dimensional human breast cultures. Both immortalized and primary human breast progenitors were analyzed. We report on the functional ability of those proteins of the mammary gland that maintain quiescence, maintain the progenitor state, and guide progenitor differentiation towards myoepithelial and luminal lineages.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Glándulas Mamarias Humanas/citología , Glándulas Mamarias Humanas/fisiología , Análisis por Matrices de Proteínas/métodos , Proteoma/metabolismo , Células Madre/citología , Células Madre/fisiología , Diferenciación Celular , Separación Celular/métodos , Supervivencia Celular , Células Cultivadas , Humanos
12.
Cancer Res ; 68(5): 1378-87, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18316601

RESUMEN

A crucial step in human breast cancer progression is the acquisition of invasiveness. There is a distinct lack of human cell culture models to study the transition from preinvasive to invasive phenotype as it may occur "spontaneously" in vivo. To delineate molecular alterations important for this transition, we isolated human breast epithelial cell lines that showed partial loss of tissue polarity in three-dimensional reconstituted basement membrane cultures. These cells remained noninvasive; however, unlike their nonmalignant counterparts, they exhibited a high propensity to acquire invasiveness through basement membrane in culture. The genomic aberrations and gene expression profiles of the cells in this model showed a high degree of similarity to primary breast tumor profiles. The xenograft tumors formed by the cell lines in three different microenvironments in nude mice displayed metaplastic phenotypes, including squamous and basal characteristics, with invasive cells exhibiting features of higher-grade tumors. To find functionally significant changes in transition from preinvasive to invasive phenotype, we performed attribute profile clustering analysis on the list of genes differentially expressed between preinvasive and invasive cells. We found integral membrane proteins, transcription factors, kinases, transport molecules, and chemokines to be highly represented. In addition, expression of matrix metalloproteinases MMP9, MMP13, MMP15, and MMP17 was up-regulated in the invasive cells. Using small interfering RNA-based approaches, we found these MMPs to be required for the invasive phenotype. This model provides a new tool for dissection of mechanisms by which preinvasive breast cells could acquire invasiveness in a metaplastic context.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Transformación Celular Neoplásica , Regulación Neoplásica de la Expresión Génica , Animales , Línea Celular Tumoral , Femenino , Perfilación de la Expresión Génica , Humanos , Metaplasia/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Modelos Biológicos , Invasividad Neoplásica , Metástasis de la Neoplasia
13.
Stem Cell Rev ; 3(2): 137-46, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17873346

RESUMEN

In most adult tissues there reside pools of stem and progenitor cells inside specialized microenvironments referred to as niches. The niche protects the stem cells from inappropriate expansion and directs their critical functions. Thus guided, stem cells are able to maintain tissue homeostasis throughout the ebb and flow of metabolic and physical demands encountered over a lifetime. Indeed, a pool of stem cells maintains mammary gland structure throughout development, and responds to the physiological demands associated with pregnancy. This review discusses how stem cells were identified in both human and mouse mammary glands; each requiring different techniques that were determined by differing biological needs and ethical constraints. These studies together create a robust portrait of mammary gland biology and identify the location of the stem cell niche, elucidate a developmental hierarchy, and suggest how the niche might be manipulated for therapeutic benefit.


Asunto(s)
Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/fisiología , Glándulas Mamarias Humanas/citología , Glándulas Mamarias Humanas/fisiología , Embarazo/fisiología , Células Madre/citología , Células Madre/fisiología , Animales , Discusiones Bioéticas , Femenino , Homeostasis/fisiología , Humanos , Ratones
14.
J Cell Biol ; 177(1): 87-101, 2007 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-17420292

RESUMEN

Cellular pathways that contribute to adult human mammary gland architecture and lineages have not been previously described. In this study, we identify a candidate stem cell niche in ducts and zones containing progenitor cells in lobules. Putative stem cells residing in ducts were essentially quiescent, whereas the progenitor cells in the lobules were more likely to be actively dividing. Cells from ducts and lobules collected under the microscope were functionally characterized by colony formation on tissue culture plastic, mammosphere formation in suspension culture, and morphogenesis in laminin-rich extracellular matrix gels. Staining for the lineage markers keratins K14 and K19 further revealed multipotent cells in the stem cell zone and three lineage-restricted cell types outside this zone. Multiparameter cell sorting and functional characterization with reference to anatomical sites in situ confirmed this pattern. The proposal that the four cell types are indeed constituents of an as of yet undescribed stem cell hierarchy was assessed in long-term cultures in which senescence was bypassed. These findings identify an adult human breast ductal stem cell activity and its earliest descendants.


Asunto(s)
Mama/citología , Linaje de la Célula , Células Madre/clasificación , Biomarcadores/metabolismo , Mama/metabolismo , Diferenciación Celular , Línea Celular , Femenino , Humanos , Queratinas/metabolismo , Glándulas Mamarias Humanas/citología , Proteínas Oncogénicas Virales/genética , Proteínas E7 de Papillomavirus , Proteínas Represoras/genética , Células Madre/metabolismo , Transducción Genética
15.
Mol Oncol ; 1(1): 84-96, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18516279

RESUMEN

3D cell cultures are rapidly becoming the method of choice for the physiologically relevant modeling of many aspects of non-malignant and malignant cell behavior ex vivo. Nevertheless, only a limited number of distinct cell types have been evaluated in this assay to date. Here we report the first large scale comparison of the transcriptional profiles and 3D cell culture phenotypes of a substantial panel of human breast cancer cell lines. Each cell line adopts a colony morphology of one of four main classes in 3D culture. These morphologies reflect, at least in part, the underlying gene expression profile and protein expression patterns of the cell lines, and distinct morphologies were also associated with tumor cell invasiveness and with cell lines originating from metastases. We further demonstrate that consistent differences in genes encoding signal transduction proteins emerge when even tumor cells are cultured in 3D microenvironments.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/biosíntesis , Transducción de Señal , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Femenino , Humanos , Invasividad Neoplásica , Metástasis de la Neoplasia
16.
Clin Cancer Res ; 11(19 Pt 1): 6872-9, 2005 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16203777

RESUMEN

PURPOSE: Microarray studies have linked Annexin A8 RNA expression to a "basal cell-like" subset of breast cancers, including BRCA1-related cancers, that are characterized by cytokeratin 5 (CK5) and CK17 expression and show poor prognosis. We assessed Annexin A8's contribution to the overall prognosis and its expression in normal, benign, and cancerous tissue and addressed Annexin A8's physiologic role in the mammary gland. EXPERIMENTAL DESIGN: Using microarrays and reverse transcription-PCR, the Annexin A8 expression was studied during mouse mammary gland development and in isolated mammary structures. Reverse transcription-PCR on cultured human luminal and basal cells, along with immunocytochemistry on normal and benign breast tissues, was used for cellular localization. Annexin A8's prognostic relevance and its coexpression with CK5 were assessed on tissue arrays of 1,631 cases of invasive breast cancer. Coexpression was further evaluated on a small cohort of 14 BRCA1-related breast cancers. RESULTS: Annexin A8 was up-regulated during mouse mammary gland involution and in pubertal ductal epithelium. Annexin A8 showed preferred expression in cultured basal cells but predominant luminal expression in normal human breast tissue in vivo. Hyperplasias and in situ carcinomas showed a strong staining of basal cells. Annexin A8 expression was significantly associated with grade (P < 0.0001), CK5 (P < 0.0001), and estrogen receptor status (P < 0.0001); 85.7% BRCA1-related breast tumors coexpressed Annexin A8 and CK5. CONCLUSION: Annexin A8 is involved in mouse mammary gland involution. In humans, it is a luminally expressed protein with basal expression in cell culture and in hyperplasia/ductal carcinoma in situ. Expression in invasive breast carcinomas has a significant effect on survival (P = 0.03) but is not independent of grade or CK5.


Asunto(s)
Anexinas/biosíntesis , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/mortalidad , Regulación Neoplásica de la Expresión Génica , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Regulación hacia Arriba , Animales , Apoptosis , Carcinoma/metabolismo , Carcinoma/patología , Carcinoma Intraductal no Infiltrante/patología , Estudios de Cohortes , Femenino , Genes BRCA1 , Humanos , Inmunohistoquímica , Queratinas/biosíntesis , Ratones , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Oligonucleótidos/química , Fenotipo , Reacción en Cadena de la Polimerasa , Pronóstico , ARN/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Resultado del Tratamiento
17.
Breast Cancer Res ; 7(5): 190-7, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16168137

RESUMEN

The mammary gland consists of an extensively branched ductal network contained within a distinctive basement membrane and encompassed by a stromal compartment. During lactation, production of milk depends on the action of the two epithelial cell types that make up the ductal network: luminal cells, which secrete the milk components into the ductal lumen; and myoepithelial cells, which contract to aid in the ejection of milk. There is increasing evidence that the myoepithelial cells also play a key role in the organizational development of the mammary gland, and that the loss and/or change of myoepithelial cell function is a key step in the development of breast cancer. In this review we briefly address the characteristics of breast myoepithelial cells from human breast and mouse mammary gland, how they function in normal mammary gland development, and their recently appreciated role in tumor suppression.


Asunto(s)
Neoplasias de la Mama/patología , Células Epiteliales/fisiología , Animales , Mama/citología , Mama/fisiología , Neoplasias de la Mama/fisiopatología , Neoplasias de la Mama/prevención & control , Polaridad Celular , Células Epiteliales/citología , Femenino , Humanos , Ratones , Valores de Referencia
18.
J Cell Sci ; 118(Pt 6): 1321-30, 2005 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15741234

RESUMEN

Nuclear organization, such as the formation of specific nuclear subdomains, is generally thought to be involved in the control of cellular phenotype; however, there are relatively few specific examples of how mammalian nuclei organize during radical changes in phenotype, such as those occurring during differentiation and growth arrest. Using human mammary epithelial cells in which growth arrest is essential for morphological differentiation, we show that the arrest of cell proliferation is accompanied by a reorganization of the telomere-associated protein, TIN2, into one to three large nuclear subdomains. The large TIN2 domains do not contain telomeres and occur concomitant with the continued presence of TIN2 at telomeres. The TIN2 domains were sensitive to DNase, but not RNase, occurred frequently, but not exclusively near nucleoli, and overlapped often with dense domains containing heterochromatin protein 1gamma. Expression of truncated forms of TIN2 simultaneously prevented the formation of TIN2 domains and relaxed the stringent morphogenesis-induced growth arrest in human mammary epithelial cells. Here we show that a novel extra-telomeric organization of TIN2 is associated with the control of cell proliferation and identify TIN2 as an important regulator of mammary epithelial differentiation.


Asunto(s)
Mama/citología , Núcleo Celular/metabolismo , Células Epiteliales/citología , Proteínas de Unión a Telómeros/metabolismo , Western Blotting , Bromodesoxiuridina/farmacología , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Desoxirribonucleasa I/metabolismo , Desoxirribonucleasas/metabolismo , Células Epiteliales/metabolismo , Heterocromatina/química , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Antígeno Ki-67/biosíntesis , Microscopía Fluorescente , Fenotipo , Estructura Terciaria de Proteína , Retroviridae/genética , Ribonucleasa Pancreática/metabolismo , Ribonucleasas/metabolismo , Telómero/metabolismo , Telómero/ultraestructura
19.
J Mammary Gland Biol Neoplasia ; 10(3): 261-72, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16807805

RESUMEN

The human breast epithelium is a branching ductal system composed of an inner layer of polarized luminal epithelial cells and an outer layer of myoepithelial cells that terminate in distally located terminal duct lobular units (TDLUs). While the luminal epithelial cell has received the most attention as the functionally active milk-producing cell and as the most likely target cell for carcinogenesis, attention on myoepithelial cells has begun to evolve with the recognition that these cells play an active part in branching morphogenesis and tumor suppression. A major question that has been the subject of investigation pertains to how the luminal epithelial and myoepithelial lineages are related and precisely how they arise from a common putative stem cell population within the breast. Equally important is the question of how heterotypic signaling occurs between luminal epithelial and surrounding myoepithelial cells in normal breast morphogenesis and neoplasia. In this review we discuss data from our laboratories and from others regarding the cellular origin of human myoepithelial cells, their function in maintaining tissue polarity in the normal breast, and their role during neoplasia.


Asunto(s)
Neoplasias de la Mama/metabolismo , Transformación Celular Neoplásica/metabolismo , Células Epiteliales/metabolismo , Glándulas Mamarias Humanas/crecimiento & desarrollo , Glándulas Mamarias Humanas/metabolismo , Morfogénesis/fisiología , Células Madre/metabolismo , Animales , Biomarcadores de Tumor/biosíntesis , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Polaridad Celular/fisiología , Células Epiteliales/citología , Femenino , Humanos , Glándulas Mamarias Humanas/citología , Células Madre/citología
20.
Lab Invest ; 83(3): 387-96, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12649339

RESUMEN

A human tumor xenograft (L56Br-X1) was established from a breast cancer axillary lymph node metastasis of a 53-year-old woman with a BRCA1 germ-line nonsense mutation (1806C>T; Q563X), and a cell line (L56Br-C1) was subsequently derived from the xenograft. The xenograft carries only the mutant BRCA1 allele and expresses mutant BRCA1 mRNA but no BRCA1 protein as determined by immunoprecipitation or Western blotting. The primary tumor, lymph node metastasis, and xenograft were hypodiploid by DNA flow cytometry, whereas the cell line displayed an aneuploidy apparently developed via polyploidization. Cytogenetic analysis, spectral karyotyping, and comparative genomic hybridization of the cell line revealed a highly complex karyotype with numerous unbalanced translocations. The xenograft and cell line had retained a somatic TP53 missense mutation (S215I) originating from the primary tumors, as well as a lack of immunohistochemically detectable expression of steroid hormone receptors, epidermal growth factor receptor, human epidermal growth factor receptor 2 (HER-2), and keratin 8. Global gene expression analysis by cDNA microarrays supported a correlation between the expression profiles of the primary tumor, lymph node metastasis, xenograft, and cell line. We conclude that L56Br-X1 and L56Br-C1 are useful model systems for studies of the pathogenesis and new therapeutic modalities of BRCA1-induced human breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Carcinoma Ductal de Mama/genética , Genes BRCA1 , Mutación de Línea Germinal/genética , Heterocigoto , Aneuploidia , Biomarcadores de Tumor/análisis , Neoplasias de la Mama/química , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/química , Carcinoma Ductal de Mama/secundario , Codón sin Sentido , ADN de Neoplasias/análisis , Femenino , Citometría de Flujo , Humanos , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Metástasis Linfática/genética , Metástasis Linfática/patología , Persona de Mediana Edad , Hibridación de Ácido Nucleico , Cariotipificación Espectral , Translocación Genética , Trasplante Heterólogo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA