Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Cell ; 183(2): 395-410.e19, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33007268

RESUMEN

Collective metastasis is defined as the cohesive migration and metastasis of multicellular tumor cell clusters. Disrupting various cell adhesion genes markedly reduces cluster formation and colonization efficiency, yet the downstream signals transmitted by clustering remain largely unknown. Here, we use mouse and human breast cancer models to identify a collective signal generated by tumor cell clusters supporting metastatic colonization. We show that tumor cell clusters produce the growth factor epigen and concentrate it within nanolumina-intercellular compartments sealed by cell-cell junctions and lined with microvilli-like protrusions. Epigen knockdown profoundly reduces metastatic outgrowth and switches clusters from a proliferative to a collective migratory state. Tumor cell clusters from basal-like 2, but not mesenchymal-like, triple-negative breast cancer cell lines have increased epigen expression, sealed nanolumina, and impaired outgrowth upon nanolumenal junction disruption. We propose that nanolumenal signaling could offer a therapeutic target for aggressive metastatic breast cancers.


Asunto(s)
Neoplasias de la Mama/fisiopatología , Uniones Intercelulares/patología , Metástasis de la Neoplasia/fisiopatología , Animales , Adhesión Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Epigen/metabolismo , Transición Epitelial-Mesenquimal/genética , Humanos , Ratones , Células Neoplásicas Circulantes/patología , Transducción de Señal/fisiología , Neoplasias de la Mama Triple Negativas/patología
2.
Cell ; 155(7): 1639-51, 2013 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-24332913

RESUMEN

Carcinomas typically invade as a cohesive multicellular unit, a process termed collective invasion. It remains unclear how different subpopulations of cancer cells contribute to this process. We developed three-dimensional (3D) organoid assays to identify the most invasive cancer cells in primary breast tumors. Collective invasion was led by specialized cancer cells that were defined by their expression of basal epithelial genes, such as cytokeratin-14 (K14) and p63. Furthermore, K14+ cells led collective invasion in the major human breast cancer subtypes. Importantly, luminal cancer cells were observed to convert phenotypically to invasive leaders following induction of basal epithelial genes. Although only a minority of cells within luminal tumors expressed basal epithelial genes, knockdown of either K14 or p63 was sufficient to block collective invasion. Our data reveal that heterotypic interactions between epithelial subpopulations are critical to collective invasion. We suggest that targeting the basal invasive program could limit metastatic progression.


Asunto(s)
Neoplasias de la Mama/patología , Invasividad Neoplásica , Animales , Neoplasias de la Mama/metabolismo , Técnicas de Cultivo de Célula , Células Cultivadas , Modelos Animales de Enfermedad , Células Epiteliales/patología , Humanos , Queratina-14/genética , Queratina-14/metabolismo , Neoplasias Pulmonares/secundario , Ratones , Organoides/patología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
3.
Proc Natl Acad Sci U S A ; 120(10): e2214888120, 2023 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-36853945

RESUMEN

Necrosis in the tumor interior is a common feature of aggressive cancers that is associated with poor clinical prognosis and the development of metastasis. How the necrotic core promotes metastasis remains unclear. Here, we report that emergence of necrosis inside the tumor is correlated temporally with increased tumor dissemination in a rat breast cancer model and in human breast cancer patients. By performing spatially focused transcriptional profiling, we identified angiopoietin-like 7 (Angptl7) as a tumor-specific factor localized to the perinecrotic zone. Functional studies showed that Angptl7 loss normalizes central necrosis, perinecrotic dilated vessels, metastasis, and reduces circulating tumor cell counts to nearly zero. Mechanistically, Angptl7 promotes vascular permeability and supports vascular remodeling in the perinecrotic zone. Taken together, these findings show that breast tumors actively produce factors controlling central necrosis formation and metastatic dissemination from the tumor core.


Asunto(s)
Neoplasias de la Mama , Neoplasias Mamarias Animales , Células Neoplásicas Circulantes , Animales , Femenino , Humanos , Ratas , Proteína 7 Similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina , Angiopoyetinas/genética , Neoplasias de la Mama/genética , Necrosis
4.
PLoS Comput Biol ; 16(1): e1007464, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31961880

RESUMEN

For women with access to healthcare and early detection, breast cancer deaths are caused primarily by metastasis rather than growth of the primary tumor. Metastasis has been difficult to study because it happens deep in the body, occurs over years, and involves a small fraction of cells from the primary tumor. Furthermore, within-tumor heterogeneity relevant to metastasis can also lead to therapy failures and is obscured by studies of bulk tissue. Here we exploit heterogeneity to identify molecular mechanisms of metastasis. We use "organoids", groups of hundreds of tumor cells taken from a patient and grown in the lab, to probe tumor heterogeneity, with potentially thousands of organoids generated from a single tumor. We show that organoids have the character of biological replicates: within-tumor and between-tumor variation are of similar magnitude. We develop new methods based on population genetics and variance components models to build between-tumor and within-tumor statistical tests, using organoids analogously to large sibships and vastly amplifying the test power. We show great efficiency for tests based on the organoids with the most extreme phenotypes and potential cost savings from pooled tests of the extreme tails, with organoids generated from hundreds of tumors having power predicted to be similar to bulk tests of hundreds of thousands of tumors. We apply these methods to an association test for molecular correlates of invasion, using a novel quantitative invasion phenotype calculated as the spectral power of the organoid boundary. These new approaches combine to show a strong association between invasion and protein expression of Keratin 14, a known biomarker for poor prognosis, with p = 2 × 10-45 for within-tumor tests of individual organoids and p < 10-6 for pooled tests of extreme tails. Future studies using these methods could lead to discoveries of new classes of cancer targets and development of corresponding therapeutics. All data and methods are available under an open source license at https://github.com/baderzone/invasion_2019.


Asunto(s)
Neoplasias de la Mama , Invasividad Neoplásica , Metástasis de la Neoplasia , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/clasificación , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Biología Computacional , Femenino , Humanos , Queratina-14/metabolismo , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Organoides/metabolismo , Organoides/patología , Células Tumorales Cultivadas
5.
J Mammary Gland Biol Neoplasia ; 25(4): 337-350, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33106923

RESUMEN

Tumor organoids mimic the architecture and heterogeneity of in vivo tumors and enable studies of collective interactions between tumor cells as well as with their surrounding microenvironment. Although tumor organoids hold significant promise as cancer models, they are also more costly and labor-intensive to cultivate than traditional 2D cell culture. We sought to identify critical factors regulating organoid growth ex vivo, and to use these observations to develop a more efficient organoid expansion method. Using time-lapse imaging of mouse mammary tumor organoids in 3D culture, we observed that outgrowth potential varies non-linearly with initial organoid size. Maximal outgrowth occurred in organoids with a starting size between ~10 to 1000 cells. Based on these observations, we developed a suspension culture method that maintains organoids in the ideal size range, enabling expansion from 1 million to over 100 million cells in less than 2 weeks and less than 3 hours of hands-on time. Our method facilitates the rapid, cost-effective expansion of organoids for CRISPR based studies and other assays requiring a large amount of organoid starting material.


Asunto(s)
Neoplasias de la Mama/patología , Técnicas de Cultivo de Célula/métodos , Organoides/patología , Esferoides Celulares/patología , Animales , Neoplasias de la Mama/genética , Sistemas CRISPR-Cas/genética , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Microscopía Intravital , Ratones , Imagen de Lapso de Tiempo , Microambiente Tumoral/genética
6.
Proc Natl Acad Sci U S A ; 113(7): E854-63, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26831077

RESUMEN

Recent genomic studies challenge the conventional model that each metastasis must arise from a single tumor cell and instead reveal that metastases can be composed of multiple genetically distinct clones. These intriguing observations raise the question: How do polyclonal metastases emerge from the primary tumor? In this study, we used multicolor lineage tracing to demonstrate that polyclonal seeding by cell clusters is a frequent mechanism in a common mouse model of breast cancer, accounting for >90% of metastases. We directly observed multicolored tumor cell clusters across major stages of metastasis, including collective invasion, local dissemination, intravascular emboli, circulating tumor cell clusters, and micrometastases. Experimentally aggregating tumor cells into clusters induced a >15-fold increase in colony formation ex vivo and a >100-fold increase in metastasis formation in vivo. Intriguingly, locally disseminated clusters, circulating tumor cell clusters, and lung micrometastases frequently expressed the epithelial cytoskeletal protein, keratin 14 (K14). RNA-seq analysis revealed that K14(+) cells were enriched for desmosome and hemidesmosome adhesion complex genes, and were depleted for MHC class II genes. Depletion of K14 expression abrogated distant metastases and disrupted expression of multiple metastasis effectors, including Tenascin C (Tnc), Jagged1 (Jag1), and Epiregulin (Ereg). Taken together, our findings reveal K14 as a key regulator of metastasis and establish the concept that K14(+) epithelial tumor cell clusters disseminate collectively to colonize distant organs.


Asunto(s)
Neoplasias de la Mama/patología , Modelos Animales de Enfermedad , Queratina-14/genética , Metástasis de la Neoplasia/genética , Animales , Neoplasias de la Mama/genética , Humanos , Ratones
7.
Proc Natl Acad Sci U S A ; 109(39): E2595-604, 2012 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-22923691

RESUMEN

Breast cancer progression involves genetic changes and changes in the extracellular matrix (ECM). To test the importance of the ECM in tumor cell dissemination, we cultured epithelium from primary human breast carcinomas in different ECM gels. We used basement membrane gels to model the normal microenvironment and collagen I to model the stromal ECM. In basement membrane gels, malignant epithelium either was indolent or grew collectively, without protrusions. In collagen I, epithelium from the same tumor invaded with protrusions and disseminated cells. Importantly, collagen I induced a similar initial response of protrusions and dissemination in both normal and malignant mammary epithelium. However, dissemination of normal cells into collagen I was transient and ceased as laminin 111 localized to the basal surface, whereas dissemination of carcinoma cells was sustained throughout culture, and laminin 111 was not detected. Despite the large impact of ECM on migration strategy, transcriptome analysis of our 3D cultures revealed few ECM-dependent changes in RNA expression. However, we observed many differences between normal and malignant epithelium, including reduced expression of cell-adhesion genes in tumors. Therefore, we tested whether deletion of an adhesion gene could induce sustained dissemination of nontransformed cells into collagen I. We found that deletion of P-cadherin was sufficient for sustained dissemination, but exclusively into collagen I. Our data reveal that metastatic tumors preferentially disseminate in specific ECM microenvironments. Furthermore, these data suggest that breaks in the basement membrane could induce invasion and dissemination via the resulting direct contact between cancer cells and collagen I.


Asunto(s)
Neoplasias de la Mama , Movimiento Celular , Regulación Neoplásica de la Expresión Génica , Neoplasias Mamarias Animales , Microambiente Tumoral , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Femenino , Humanos , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Ratones , Invasividad Neoplásica
8.
J Cell Sci ; 125(Pt 11): 2638-54, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22344263

RESUMEN

Normal mammary morphogenesis involves transitions between simple and multilayered epithelial organizations. We used electron microscopy and molecular markers to determine whether intercellular junctions and apico-basal polarity were maintained in the multilayered epithelium. We found that multilayered elongating ducts had polarized apical and basal tissue surfaces both in three-dimensional culture and in vivo. However, individual cells were only polarized on surfaces in contact with the lumen or extracellular matrix. The basolateral marker scribble and the apical marker atypical protein kinase C zeta localized to all interior cell membranes, whereas PAR3 displayed a cytoplasmic localization, suggesting that the apico-basal polarity was incomplete. Despite membrane localization of E-cadherin and ß-catenin, we did not observe a defined zonula adherens connecting interior cells. Instead, interior cells were connected through desmosomes and exhibited complex interdigitating membrane protrusions. Single-cell labeling revealed that individual cells were both protrusive and migratory within the epithelial multilayer. Inhibition of Rho kinase (ROCK) further reduced intercellular adhesion on apical and lateral surfaces but did not disrupt basal tissue organization. Following morphogenesis, segregated membrane domains were re-established and junctional complexes re-formed. We observed similar epithelial organization during mammary morphogenesis in organotypic culture and in vivo. We conclude that mammary epithelial morphogenesis involves a reversible, spatially limited, reduction in polarity and intercellular junctions and active individualistic cell migration. Our data suggest that reductions in polarity and adhesion during breast cancer progression might reflect partial recapitulation of a normal developmental program.


Asunto(s)
Movimiento Celular , Células Epiteliales/citología , Epitelio/metabolismo , Glándulas Mamarias Animales/citología , Amidas/farmacología , Animales , Comunicación Celular/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Movimiento Celular/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Extensiones de la Superficie Celular/metabolismo , Extensiones de la Superficie Celular/ultraestructura , Colágeno , Desmosomas/efectos de los fármacos , Desmosomas/metabolismo , Desmosomas/ultraestructura , Combinación de Medicamentos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Células Epiteliales/ultraestructura , Epitelio/efectos de los fármacos , Epitelio/ultraestructura , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Femenino , Laminina , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/ultraestructura , Ratones , Modelos Biológicos , Morfogénesis/efectos de los fármacos , Quinasa de Cadena Ligera de Miosina/metabolismo , Organoides/citología , Organoides/efectos de los fármacos , Organoides/ultraestructura , Proteoglicanos , Piridinas/farmacología , Uniones Estrechas/metabolismo , Uniones Estrechas/ultraestructura , Técnicas de Cultivo de Tejidos , Proteínas de Unión al GTP rac/metabolismo , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
9.
Annu Rev Pathol ; 18: 231-256, 2023 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-36207009

RESUMEN

Metastatic dissemination has lethal consequences for cancer patients. Accruing evidence supports the hypothesis that tumor cells can migrate and metastasize as clusters of cells while maintaining contacts with one another. Collective metastasis enables tumor cells to colonize secondary sites more efficiently, resist cell death, and evade the immune system. On the other hand, tumor cell clusters face unique challenges for dissemination particularly during systemic dissemination. Here, we review recent progress toward understanding how tumor cell clusters overcome these disadvantages as well as mechanisms they utilize to gain advantages throughout the metastatic process. We consider useful models for studying collective metastasis and reflect on how the study of collective metastasis suggests new opportunities for eradicating and preventing metastatic disease.


Asunto(s)
Neoplasias , Humanos , Movimiento Celular
10.
Cell Metab ; 5(2): 115-28, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17276354

RESUMEN

In an effort to identify novel candidate regulators of adipogenesis, gene profiling of differentiating 3T3-L1 preadipocytes was analyzed using a novel algorithm. We report here the characterization of xanthine oxidoreductase (XOR) as a novel regulator of adipogenesis. XOR lies downstream of C/EBPbeta and upstream of PPARgamma, in the cascade of factors that control adipogenesis, and it regulates PPARgamma activity. In vitro, knockdown of XOR inhibits adipogenesis and PPARgamma activity while constitutive overexpression increases activity of the PPARgamma receptor in both adipocytes and preadipocytes. In vivo, XOR -/- mice demonstrate 50% reduction in adipose mass versus wild-type littermates while obese ob/ob mice exhibit increased concentrations of XOR mRNA and urate in the adipose tissue. We propose that XOR is a novel regulator of adipogenesis and of PPARgamma activity and essential for the regulation of fat accretion. Our results identify XOR as a potential therapeutic target for metabolic abnormalities beyond hyperuricemia.


Asunto(s)
Adipogénesis , PPAR gamma/genética , PPAR gamma/metabolismo , Xantina Deshidrogenasa/metabolismo , Células 3T3-L1 , Adipocitos/citología , Adipocitos/efectos de los fármacos , Adipocitos/enzimología , Adipogénesis/efectos de los fármacos , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/enzimología , Adiposidad/efectos de los fármacos , Animales , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Perfilación de la Expresión Génica , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Tamaño de los Órganos/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Rosiglitazona , Tiazolidinedionas/farmacología , Xantina Deshidrogenasa/deficiencia , Xantina Deshidrogenasa/genética
11.
NPJ Breast Cancer ; 7(1): 42, 2021 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-33863924

RESUMEN

Angiogenesis is a critical component of breast cancer development, and identification of imaging-based angiogenesis assays has prognostic and treatment implications. We evaluated the association of semi-quantitative kinetic and radiomic breast cancer features on dynamic contrast-enhanced (DCE)-MRI with microvessel density (MVD), a marker for angiogenesis. Invasive breast cancer kinetic features (initial peak percent enhancement [PE], signal enhancement ratio [SER], functional tumor volume [FTV], and washout fraction [WF]), radiomics features (108 total features reflecting tumor morphology, signal intensity, and texture), and MVD (by histologic CD31 immunostaining) were measured in 27 patients (1/2016-7/2017). Lesions with high MVD levels demonstrated higher peak SER than lesions with low MVD (mean: 1.94 vs. 1.61, area under the receiver operating characteristic curve [AUC] = 0.79, p = 0.009) and higher WF (mean: 50.6% vs. 22.5%, AUC = 0.87, p = 0.001). Several radiomics texture features were also promising for predicting increased MVD (maximum AUC = 0.84, p = 0.002). Our study suggests DCE-MRI can non-invasively assess breast cancer angiogenesis, which could stratify biology and optimize treatments.

12.
Nat Protoc ; 15(8): 2413-2442, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32690957

RESUMEN

Cancer invasion and metastasis are challenging to study in vivo since they occur deep inside the body over extended time periods. Organotypic 3D culture of fresh tumor tissue enables convenient real-time imaging, genetic and microenvironmental manipulation and molecular analysis. Here, we provide detailed protocols to isolate and culture heterogenous organoids from murine and human primary and metastatic site tumors. The time required to isolate organoids can vary based on the tissue and organ type but typically takes <7 h. We describe a suite of assays that model specific aspects of metastasis, including proliferation, survival, invasion, dissemination and colony formation. We also specify comprehensive protocols for downstream applications of organotypic cultures that will allow users to (i) test the role of specific genes in regulating various cellular processes, (ii) distinguish the contributions of several microenvironmental factors and (iii) test the effects of novel therapeutics.


Asunto(s)
Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , Técnicas de Cultivo de Tejidos/métodos , Animales , Humanos , Ratones , Metástasis de la Neoplasia
13.
Dev Cell ; 47(5): 539-540, 2018 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-30513297

RESUMEN

Stem cells can enter a reversible cell-cycle arrest state termed quiescence. In this issue of Developmental Cell, Fu et al. report that the transcription factor FoxP1 is necessary for postnatal mammary gland development and relieves Tspan8-dependent quiescence in basal mammary stem cells.


Asunto(s)
Células Madre/citología , Puntos de Control del Ciclo Celular , División Celular , Proliferación Celular , Morfogénesis
14.
Dev Cell ; 40(4): 328-330, 2017 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-28245919

RESUMEN

The properties of stem cells that participate in mammary gland branching morphogenesis remain contested. Reporting in Nature, Scheele et al. (2017) establish a model for post-pubertal mammary branching morphogenesis in which position-dependent, lineage-restricted stem cells undergo cell mixing in order to contribute to long-term growth.


Asunto(s)
Glándulas Mamarias Animales , Morfogénesis , Animales , Células Epiteliales , Células Madre
15.
Science ; 352(6282): 167-9, 2016 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-27124449

RESUMEN

Despite decades of study, there are still many unanswered questions about metastasis, the process by which a localized cancer becomes a systemic disease. One of these questions is the nature of the tumor cells that give rise to metastases. Although conventional models suggest that metastases are seeded by single cells from the primary tumor, there is growing evidence that seeding requires the collective action of tumor cells traveling together in clusters. Here, we review this evidence, which comes from analysis of both experimental models and patient samples. We present a model of metastatic dissemination that highlights the activities of clusters of tumor cells that retain and require their epithelial properties.


Asunto(s)
Modelos Biológicos , Metástasis de la Neoplasia/patología , Células Neoplásicas Circulantes/patología , Animales , Células Epiteliales/patología , Humanos , Ratones , Siembra Neoplásica , Neoplasias Experimentales/patología
16.
Methods Mol Biol ; 1189: 135-62, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25245692

RESUMEN

Epithelia are fundamental tissues that line cavities, glands, and outer body surfaces. We use three-dimensional (3D) embedded culture of primary murine mammary epithelial ducts, called "organoids," to recapitulate in days in culture epithelial programs that occur over weeks deep within the body. Modulating the composition of the extracellular matrix (ECM) allows us to model cell- and tissue-level behaviors observed in normal development, such as branching morphogenesis, and in cancer, such as invasion and dissemination. Here, we describe a collection of protocols for 3D culture of mammary organoids in different ECMs and for immunofluorescence staining of 3D culture samples and mammary gland tissue sections. We illustrate expected phenotypic outcomes of each assay and provide troubleshooting tips for commonly encountered technical problems.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Epiteliales/citología , Glándulas Mamarias Animales/crecimiento & desarrollo , Morfogénesis , Animales , Bioensayo , Separación Celular , Forma de la Célula/efectos de los fármacos , Colágeno/farmacología , Colágeno Tipo I/farmacología , Combinación de Medicamentos , Células Epiteliales/efectos de los fármacos , Femenino , Técnica del Anticuerpo Fluorescente , Laminina/farmacología , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/efectos de los fármacos , Ratones , Morfogénesis/efectos de los fármacos , Organoides/citología , Organoides/efectos de los fármacos , Fenotipo , Proteoglicanos/farmacología , Ratas , Albúmina Sérica Bovina/metabolismo , Coloración y Etiquetado
18.
Curr Opin Cell Biol ; 30: 99-111, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25137487

RESUMEN

Metastasis begins when tumors invade into surrounding tissues. In breast cancer, the study of cell interactions has provided fundamental insights into this complex process. Powerful intravital and 3D organoid culture systems have emerged that enable biologists to model the complexity of cell interactions during cancer invasion in real-time. Recent studies utilizing these techniques reveal distinct mechanisms through which multiple cancer cell and stromal cell subpopulations interact, including paracrine signaling, direct cell-cell adhesion, and remodeling of the extracellular matrix. Three cell interaction mechanisms have emerged to explain how breast tumors become invasive: epithelial-mesenchymal transition, collective invasion, and the macrophage-tumor cell feedback loop. Future work is needed to distinguish whether these mechanisms are mutually exclusive or whether they cooperate to drive metastasis.


Asunto(s)
Neoplasias de la Mama/patología , Comunicación Celular , Animales , Neoplasias de la Mama/metabolismo , Transición Epitelial-Mesenquimal , Matriz Extracelular/metabolismo , Humanos , Invasividad Neoplásica , Metástasis de la Neoplasia
20.
Curr Opin Cell Biol ; 22(5): 640-50, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20832275

RESUMEN

The cellular mechanisms driving mammalian epithelial morphogenesis are of significant fundamental and practical interest. Historically, these processes have been difficult to study directly, owing to the opacity and relative inaccessibility of mammalian tissues. Recent experimental advances in timelapse imaging and in 3D organotypic culture have enabled direct observation of epithelial morphogenesis. In the mammary gland, branching morphogenesis is observed to proceed through a novel form of collective epithelial migration. The active unit of morphogenesis is a multilayered epithelium with reduced apico-basal polarity, within which cells rearranged vigorously. From within this multilayered state, new ducts initiate and elongate into the matrix without leading cellular extensions or dedicated leaders. We discuss the implications of these findings on our understanding of epithelial morphogenesis in other organs and in cancer progression.


Asunto(s)
Epitelio/crecimiento & desarrollo , Morfogénesis , Neoplasias , Animales , Humanos , Modelos Biológicos , Neoplasias/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA