Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 4866, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38849373

RESUMEN

Dense and aligned Collagen I fibers are associated with collective cancer invasion led by protrusive tumor cells, leader cells. In some breast tumors, a population of cancer cells (basal-like cells) maintain several epithelial characteristics and express the myoepithelial/basal cell marker Keratin 14 (K14). Emergence of leader cells and K14 expression are regarded as interconnected events triggered by Collagen I, however the underlying mechanisms remain unknown. Using breast carcinoma organoids, we show that Collagen I drives a force-dependent loop, specifically in basal-like cancer cells. The feed-forward loop is centered around the mechanotransducer Yap and independent of K14 expression. Yap promotes a transcriptional program that enhances Collagen I alignment and tension, which further activates Yap. Active Yap is detected in invading breast cancer cells in patients and required for collective invasion in 3D Collagen I and in the mammary fat pad of mice. Our work uncovers an essential function for Yap in leader cell selection during collective cancer invasion.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Neoplasias de la Mama , Colágeno Tipo I , Mecanotransducción Celular , Invasividad Neoplásica , Factores de Transcripción , Proteínas Señalizadoras YAP , Animales , Femenino , Humanos , Ratones , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/genética , Línea Celular Tumoral , Colágeno Tipo I/metabolismo , Regulación Neoplásica de la Expresión Génica , Organoides/metabolismo , Organoides/patología , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Proteínas Señalizadoras YAP/metabolismo
2.
Sci Transl Med ; 16(728): eadg3840, 2024 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-38170791

RESUMEN

The extracellular matrix (ECM) is essential for cell support during homeostasis and plays a critical role in cancer. Although research often concentrates on the tumor's cellular aspect, attention is growing for the importance of the cancer-associated ECM. Biochemical and physical ECM signals affect tumor formation, invasion, metastasis, and therapy resistance. Examining the tumor microenvironment uncovers intricate ECM dysregulation and interactions with cancer and stromal cells. Anticancer therapies targeting ECM sensors and remodelers, including integrins and matrix metalloproteinases, and ECM-remodeling cells, have seen limited success. This review explores the ECM's role in cancer and discusses potential therapeutic strategies for cell-ECM interactions.


Asunto(s)
Neoplasias , Humanos , Fenómenos Biomecánicos , Neoplasias/patología , Matriz Extracelular , Integrinas , Microambiente Tumoral
3.
Methods Mol Biol ; 2608: 281-303, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36653714

RESUMEN

Cancer-derived organoids and three-dimensional (3D) extracellular matrix (ECM) are taking center stage as in vitro models to study neoplastic cell behavior, since they recapitulate the heterogeneous cellular composition of tumors and their extracellular environment. In combination with imaging and molecular/biochemical techniques, 3D organoid models have contributed substantially to our knowledge about the cellular and molecular mechanisms that regulate the growth of tumors and invasion into the surrounding tissue. We here outline a set of protocols that describe culturing of cancer-derived organoids in 3D matrices and various strategies that allow modeling of tumor growth, tumor cell penetration into basement membranes, and invasion into Collagen I-rich ECM. Furthermore, we specify protocols for subsequent handling of organoids cultured in 3D ECM for confocal microscopy and analysis of gene expression at the protein and mRNA level. Although we here use breast cancer-derived organoids, these protocols can be directly applied or adapted for organoids derived from other cancer types or healthy tissues. Thus, in addition to investigating cell behavior of multiple cancer types, the combination of protocols described here may be used to study processes such as cell differentiation and migration during homeostasis and normal development.


Asunto(s)
Neoplasias de la Mama , Matriz Extracelular , Humanos , Femenino , Matriz Extracelular/metabolismo , Colágeno Tipo I/metabolismo , Neoplasias de la Mama/patología , Membrana Basal , Organoides
4.
Oncogene ; 40(45): 6343-6353, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34584219

RESUMEN

In breast cancer the transcription factor SOX4 has been shown to be associated with poor survival, increased tumor size and metastasis formation. This has mostly been attributed to the ability of SOX4 to regulate Epithelial-to-Mesenchymal-Transition (EMT). However, SOX4 regulates target gene transcription in a context-dependent manner that is determined by the cellular and epigenetic state. In this study we have investigated the loss of SOX4 in mammary tumor development utilizing organoids derived from a PyMT genetic mouse model of breast cancer. Using CRISPR/Cas9 to abrogate SOX4 expression, we found that SOX4 is required for inhibiting differentiation by regulating a subset of genes that are highly activated in fetal mammary stem cells (fMaSC). In this way, SOX4 re-activates an oncogenic transcriptional program that is regulated in many progenitor cell-types during embryonic development. SOX4-knockout organoids are characterized by the presence of more differentiated cells that exhibit luminal or basal gene expression patterns, but lower expression of cell cycle genes. In agreement, primary tumor growth and metastatic outgrowth in the lungs are impaired in SOX4KO tumors. Finally, SOX4KO tumors show a severe loss in competitive capacity to grow out compared to SOX4-proficient cells in primary tumors. Our study identifies a novel role for SOX4 in maintaining mammary tumors in an undifferentiated and proliferative state. Therapeutic manipulation of SOX4 function could provide a novel strategy for cancer differentiation therapy, which would promote differentiation and inhibit cycling of tumor cells.


Asunto(s)
Neoplasias de la Mama/patología , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Organoides/trasplante , Factores de Transcripción SOXC/genética , Animales , Neoplasias de la Mama/genética , Sistemas CRISPR-Cas , Proteínas de Ciclo Celular/genética , Transición Epitelial-Mesenquimal , Femenino , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Neoplasias Pulmonares/genética , Ratones , Trasplante de Neoplasias , Organoides/patología
5.
J Cell Biol ; 219(10)2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32777015

RESUMEN

Progression of epithelial cancers predominantly proceeds by collective invasion of cell groups with coordinated cell-cell junctions and multicellular cytoskeletal activity. Collectively invading breast cancer cells express the gap junction protein connexin-43 (Cx43), yet whether Cx43 regulates collective invasion remains unclear. We here show that Cx43 mediates gap-junctional coupling between collectively invading breast cancer cells and, via hemichannels, adenosine nucleotide/nucleoside release into the extracellular space. Using molecular interference and rescue strategies, we identify that Cx43 hemichannel function, but not intercellular communication, induces leader cell activity and collective migration through the engagement of the adenosine receptor 1 (ADORA1) and AKT signaling. Accordingly, pharmacological inhibition of ADORA1 or AKT signaling caused leader cell collapse and halted collective invasion. ADORA1 inhibition further reduced local invasion of orthotopic mammary tumors in vivo, and joint up-regulation of Cx43 and ADORA1 in breast cancer patients correlated with decreased relapse-free survival. This identifies autocrine purinergic signaling, through Cx43 hemichannels, as a critical pathway in leader cell function and collective invasion.


Asunto(s)
Neoplasias de la Mama/genética , Conexina 43/genética , Invasividad Neoplásica/genética , Receptores Purinérgicos P1/genética , Adenosina Trifosfato/genética , Neoplasias de la Mama/patología , Comunicación Celular/genética , Línea Celular Tumoral , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Uniones Comunicantes/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Uniones Intercelulares/genética , Invasividad Neoplásica/patología , Proteínas Proto-Oncogénicas c-akt/genética , Transducción de Señal/genética
6.
Exp Cell Res ; 376(1): 86-91, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30633881

RESUMEN

Collective invasion drives the spread of multicellular cancer groups, into the normal tissue surrounding several epithelial tumors. Collective invasion recapitulates various aspects of the multicellular organization and collective migration that take place during normal development and repair. Collective migration starts with the specification of leader cells in which a polarized, migratory phenotype is established. Leader cells initiate and organize the migration of follower cells, to allow the group of cells to move as a cohesive and polarized unit. Leader-follower specification is essential for coordinated and directional collective movement. Forces exerted by cohesive cells represent key signals that dictate multicellular coordination and directionality. Physical forces originate from the contraction of the actomyosin cytoskeleton, which is linked between cells via cadherin-based cell-cell junctions. The cadherin complex senses and transduces fluctuations in forces into biochemical signals that regulate processes like cell proliferation, motility and polarity. With cadherin junctions being maintained in most collective movements the cadherin complex is ideally positioned to integrate mechanical information into the organization of collective cell migration. Here we discuss the potential roles of cadherin mechanotransduction in the diverse aspects of leader versus follower cell specification during collective migration and neoplastic invasion.


Asunto(s)
Cadherinas/genética , Movimiento Celular/genética , Polaridad Celular/genética , Mecanotransducción Celular/genética , Actomiosina/genética , Animales , Cadherinas/química , Humanos , Uniones Intercelulares/genética , Microtúbulos/genética
7.
Clin Exp Metastasis ; 34(6-7): 421-429, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28894989

RESUMEN

Breast cancer undergoes collective tissue invasion and, in experimental models, can collectively metastasize. The prevalence of collective invasion and its contribution to distant metastasis in clinical disease, however, remains poorly defined. We here scored the adipose tissue invasion of primary invasive ductal carcinoma (IDC), expressing E-cadherin, and E-cadherin negative invasive lobular carcinoma (ILC) and identified predominantly collective invasion patterns (86/86 samples) in both carcinoma types. Whereas collective invasion in IDC lesions retained adherens junctions, multicellular clusters and "Indian files" in ILC, despite the absence of adherens junctions (AJ) proteins E-cadherin and ß-catenin, retained CD44 at cell-cell contacts. By histomorphological scoring and semi-automated image analysis, we show that the extent of collective invasion into the adipose tissue correlated with decreased distant metastasis-free survival (5-year follow-up; hazard ratio: 2.32 and 2.29, respectively). Thus, collective invasion represents the predominant invasion mode in breast cancer, develops distinct junctional subtypes in IDC and ILC, and associates with distant metastasis, suggesting a critical role in systemic dissemination.


Asunto(s)
Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/patología , Carcinoma Lobular/patología , Invasividad Neoplásica/patología , Adulto , Anciano , Neoplasias de la Mama/mortalidad , Carcinoma Ductal de Mama/mortalidad , Carcinoma Lobular/mortalidad , Supervivencia sin Enfermedad , Femenino , Humanos , Estimación de Kaplan-Meier , Persona de Mediana Edad
8.
Cell Commun Adhes ; 18(5): 104-16, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22142338

RESUMEN

Crosstalk between gap junction intracellular communication (GJIC), STAT5 and OCT-1 in gap junction (GJ)-dependent ß-casein expression was investigated. CID-9 mammary cells plated with prolactin on non-adherent substratum (poly-HEMA) expressed ß-casein independent of STAT5 only in the presence of the GJIC inducer, cAMP. Nuclear STAT5 levels were not detectable. By contrast, cells on EHS-drip expressed ß-casein in a STAT5-dependent manner and nuclear STAT5 levels were up-regulated. A 75 kDa OCT-1 isoform was detected in conditions that induced ß-casein expression regardless of substratum. Interestingly, 40 and 28 kDa OCT-1 isoforms were induced in cells on polyHEMA with cAMP. Electrophoretic mobility shift assays (EMSA) for OCT-1 revealed two band shifts in cells on polyHEMA with cAMP and on EHS-drip, which were repressed by the GJIC inhibitor, 18α-GA. These studies demonstrated that mammary cells on polyHEMA expressed ß-casein in response to prolactin in a pathway that involves GJIC and OCT-1 and is independent of STAT5 nuclear translocation.


Asunto(s)
Caseínas/metabolismo , Células Epiteliales/metabolismo , Uniones Comunicantes/metabolismo , Glándulas Mamarias Animales/citología , Factor de Transcripción STAT5/metabolismo , Transporte Activo de Núcleo Celular , Animales , Caseínas/genética , Técnicas de Cultivo de Célula , Diferenciación Celular , Línea Celular , Sondas de ADN/química , Femenino , Expresión Génica , Regulación de la Expresión Génica , Janus Quinasa 2/metabolismo , Ratones , Factor 1 de Transcripción de Unión a Octámeros/química , Factor 1 de Transcripción de Unión a Octámeros/genética , Factor 1 de Transcripción de Unión a Octámeros/metabolismo , Fosforilación , Prolactina/farmacología , Prolactina/fisiología , Unión Proteica , Receptor Cross-Talk , Transducción de Señal
9.
Integr Biol (Camb) ; 2(11-12): 568-74, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20886167

RESUMEN

Collective migration is a basic mechanism of cell translocation during morphogenesis, wound repair and cancer invasion. Collective movement requires cells to retain cell-cell contacts, exhibit group polarization with defined front-rear asymmetry, and consequently move as one multicellular unit. Depending on the cell type, morphology of the group and the tissue context, distinct mechanisms control the leading edge dynamics and guidance. Leading edge migration may either result from adhesion to ECM and contractile pulling, or from forward pushing. The leading edge consists of either one or few dedicated tip cells or a multicellular leading row that generate adhesion and traction towards the tissue substrate. Alternatively, a multicellular bud consisting of many cells protrudes collectively by proliferation and growth thereby mechanically expanding and pushing towards the tissue stroma. Each type of collective guidance engages distinct spatiotemporal molecular control and feedback towards rearward cells and the adjacent tissue microenvironment; these include intrinsic polarity mechanisms regulated by the interplay between cell-cell and cell-ECM interactions; or the heterotypic integration of stromal cells that adopt leader cell functions. We here classify molecular and mechanical mechanisms of leading function in collective cell migration during morphogenesis and wound repair and discuss how these are recapitulated during collective invasion of cancer cells.


Asunto(s)
Comunicación Celular/fisiología , Movimiento Celular/fisiología , Animales , Fenómenos Biomecánicos , Diferenciación Celular , Polaridad Celular , Matriz Extracelular/fisiología , Femenino , Humanos , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/crecimiento & desarrollo , Modelos Biológicos , Morfogénesis , Invasividad Neoplásica , Regeneración , Transducción de Señal , Cicatrización de Heridas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...