Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Methods Mol Biol ; 2748: 1-12, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38070103

RESUMEN

Isolation of live cells from the tumor microenvironment (TME) has represented a challenge, particularly from metastatic nodules that need to be identified within the entire metastatic tissue. Cherry-niche, an in vivo labelling technique, allows the isolation of all the different cell populations in the TME without needing to visually locate the metastatic cancer cell colonies. Therefore, neighboring TME cells can be isolated even from the early stages of cancer cell seeding and colonization in the metastatic tissue. Here, we show how to use Cherry-niche to identify and isolate neutrophils from the lung metastatic niche. We also provide examples of downstream analyses to characterize freshly isolated neutrophils ex vivo, such as Giemsa staining, reactive oxygen species (ROS) detection, and phagocytosis assays. Similar strategies can be used to isolate other immune and non-immune cells from the metastatic TME.


Asunto(s)
Pulmón , Microambiente Tumoral , Pulmón/patología , Neutrófilos/patología , Línea Celular Tumoral
3.
Cancer Lett ; 544: 215800, 2022 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-35803476

RESUMEN

Cancer cells thrive when embedded in a fine-tuned cellular and extracellular environment or tumour microenvironment (TME). There is a general understanding of a co-evolution between cancer cells and their surrounding TME, pointing at a functional connection between cancer cells characteristics and the perturbations induced in their surrounding tissue. However, it has never been formally proven whether this functional connection needs to be set from the start or if aggressive cancer cells always dominate their microenvironmental any point in time. This would require a dedicated experimental setting where malignant cells are challenged to grow in a different TME from the one they would naturally create. Here we generated an experimental setting where we transiently perturb the secretory profile of aggressive breast cancer cells without affecting their intrinsic growth ability, which led to the initial establishment of an atypical TME. Interestingly, even if initially tumours are formed, this atypical TME evolves to impair long term in vivo cancer growth. Using a combination of in vivo transcriptomics, protein arrays and in vitro co-cultures, we found that the atypical TME culminates in the infiltration of macrophages with STAT1high activity. These macrophages show strong anti-tumoural functions which reduce long-term tumour growth, despite lacking canonical M1 markers. Importantly, gene signatures of the mesenchymal compartment of the TME, as well as the anti-tumoural macrophages, show striking prognostic power that correlates with less aggressive human breast cancers.


Asunto(s)
Neoplasias de la Mama , Microambiente Tumoral , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Femenino , Humanos , Macrófagos/patología
4.
Cell Rep ; 39(9): 110871, 2022 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-35649380

RESUMEN

The maintenance of genome stability relies on coordinated control of origin activation and replication fork progression. How the interplay between these processes influences human genetic disease and cancer remains incompletely characterized. Here we show that mouse cells featuring Polε instability exhibit impaired genome-wide activation of DNA replication origins, in an origin-location-independent manner. Strikingly, Trp53 ablation in primary Polε hypomorphic cells increased Polε levels and origin activation and reduced DNA damage in a transcription-dependent manner. Transcriptome analysis of primary Trp53 knockout cells revealed that the TRP53-CDKN1A/P21 axis maintains appropriate levels of replication factors and CDK activity during unchallenged S phase. Loss of this control mechanism deregulates origin activation and perturbs genome-wide replication fork progression. Thus, while our data support an impaired origin activation model for genetic diseases affecting CMG formation, we propose that loss of the TRP53-CDKN1A/P21 tumor suppressor axis induces inappropriate origin activation and deregulates genome-wide fork progression.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina , ADN Polimerasa II , Replicación del ADN , Proteínas de Unión a Poli-ADP-Ribosa , Origen de Réplica , Proteína p53 Supresora de Tumor , Animales , Proteínas de Ciclo Celular/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Daño del ADN/genética , ADN Polimerasa II/genética , Replicación del ADN/genética , Ratones , Proteínas de Unión a Poli-ADP-Ribosa/genética , Fase S , Proteína p53 Supresora de Tumor/genética
6.
Nat Cancer ; 3(2): 173-187, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35221334

RESUMEN

Radiotherapy is one of the most effective approaches to achieve tumor control in cancer patients, although healthy tissue injury due to off-target radiation exposure can occur. In this study, we used a model of acute radiation injury to the lung, in the context of cancer metastasis, to understand the biological link between tissue damage and cancer progression. We exposed healthy mouse lung tissue to radiation before the induction of metastasis and observed a strong enhancement of cancer cell growth. We found that locally activated neutrophils were key drivers of the tumor-supportive preconditioning of the lung microenvironment, governed by enhanced regenerative Notch signaling. Importantly, these tissue perturbations endowed arriving cancer cells with an augmented stemness phenotype. By preventing neutrophil-dependent Notch activation, via blocking degranulation, we were able to significantly offset the radiation-enhanced metastases. This work highlights a pro-tumorigenic activity of neutrophils, which is likely linked to their tissue regenerative functions.


Asunto(s)
Neoplasias Pulmonares , Exposición a la Radiación , Animales , Humanos , Pulmón/patología , Neoplasias Pulmonares/patología , Ratones , Activación Neutrófila , Neutrófilos/patología , Microambiente Tumoral
7.
Cell Rep ; 35(6): 109119, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33979628

RESUMEN

The bone-marrow (BM) niche is the spatial environment composed by a network of multiple stromal components regulating adult hematopoiesis. We use multi-omics and computational tools to analyze multiple BM environmental compartments and decipher their mutual interactions in the context of acute myeloid leukemia (AML) xenografts. Under homeostatic conditions, we find a considerable overlap between niche populations identified using current markers. Our analysis defines eight functional clusters of genes informing on the cellular identity and function of the different subpopulations and pointing at specific stromal interrelationships. We describe how these transcriptomic profiles change during human AML development and, by using a proximity-based molecular approach, we identify early disease onset deregulated genes in the mesenchymal compartment. Finally, we analyze the BM proteomic secretome in the presence of AML and integrate it with the transcriptome to predict signaling nodes involved in niche alteration in AML.


Asunto(s)
Células de la Médula Ósea/metabolismo , Leucemia Mieloide Aguda/genética , Proteómica/métodos , Animales , Humanos , Ratones , Microambiente Tumoral
8.
Nat Protoc ; 16(2): 872-892, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33311715

RESUMEN

Understanding cell-cell interactions is critical in most, if not all, research fields in biology. Nevertheless, studying intercellular crosstalk in vivo remains a relevant challenge, due mainly to the difficulty in spatially locating the surroundings of particular cells in the tissue. Cherry-niche is a powerful new method that enables cells expressing a fluorescent protein to label their surrounding cells, facilitating their specific isolation from the whole tissue as live cells. We previously applied Cherry-niche in cancer research to study the tumor microenvironment (TME) in metastasis. Here we describe how to generate cancer cells with the ability to label their neighboring cells (within the tumor niche) by transferring a liposoluble fluorescent protein. Live niche cells can be isolated and compared with cells distant from the tumor bulk, using a variety of ex vivo approaches. As previously shown, this system has the potential to identify novel components in the TME and improve our understanding of their local interactions. Importantly, Cherry-niche can also be applied to study potential cell-cell interactions due to in vivo proximity in research fields beyond cancer. This protocol takes 2-3 weeks to generate the labeling cells and 1-2 weeks to test their labeling ability.


Asunto(s)
Comunicación Celular/fisiología , Inmunohistoquímica/métodos , Colorantes Fluorescentes/química , Humanos , Neoplasias/inmunología , Neoplasias/patología , Microambiente Tumoral/inmunología , Microambiente Tumoral/fisiología
9.
Front Oncol ; 10: 594514, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33251149

RESUMEN

Metastases are a major cause of cancer-related death and despite the fact that they have been focus of intense research over the last two decades, effective therapies for patients with distant secondary lesions are still very limited. In addition, in some tumor types metastases can grow years after the patients have been declared clinically cured, indicating that disseminated cancer cells (DCCs) persist undetected for years, even decades in a quiescent state. Clinical and experimental data highlight the importance of the immune system in shaping the fitness and behaviour of DCCs. Here, we review mechanisms of survival, quiescence and outgrowth of DCCs with a special focus on immune-regulation and we highlight the latest cutting-edge techniques for modelling the biology of DCCs in vitro and for studying the metastatic niche in vivo. We believe that a wide dissemination of those techniques will boost scientific findings towards new therapies to defeat metastatic relapses in cancer patients.

11.
Nature ; 572(7771): 603-608, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31462798

RESUMEN

Direct investigation of the early cellular changes induced by metastatic cells within the surrounding tissue remains a challenge. Here we present a system in which metastatic cancer cells release a cell-penetrating fluorescent protein, which is taken up by neighbouring cells and enables spatial identification of the local metastatic cellular environment. Using this system, tissue cells with low representation in the metastatic niche can be identified and characterized within the bulk tissue. To highlight its potential, we applied this strategy to study the cellular environment of metastatic breast cancer cells in the lung. We report the presence of cancer-associated parenchymal cells, which exhibit stem-cell-like features, expression of lung progenitor markers, multi-lineage differentiation potential and self-renewal activity. In ex vivo assays, lung epithelial cells acquire a cancer-associated parenchymal-cell-like phenotype when co-cultured with cancer cells and support their growth. These results highlight the potential of this method as a platform for new discoveries.


Asunto(s)
Linaje de la Célula , Rastreo Celular/métodos , Metástasis de la Neoplasia/patología , Células Madre Neoplásicas/patología , Tejido Parenquimatoso/patología , Coloración y Etiquetado/métodos , Nicho de Células Madre , Microambiente Tumoral , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Diferenciación Celular , Técnicas de Cocultivo , Células Epiteliales/patología , Femenino , Humanos , Proteínas Luminiscentes/análisis , Proteínas Luminiscentes/química , Proteínas Luminiscentes/metabolismo , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Masculino , Ratones , Metástasis de la Neoplasia/inmunología , Neutrófilos/patología , Organoides/patología , Nicho de Células Madre/inmunología , Microambiente Tumoral/inmunología , Proteína Fluorescente Roja
13.
Nat Commun ; 10(1): 903, 2019 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-30796225

RESUMEN

Converting carcinomas in benign oncocytomas has been suggested as a potential anti-cancer strategy. One of the oncocytoma hallmarks is the lack of respiratory complex I (CI). Here we use genetic ablation of this enzyme to induce indolence in two cancer types, and show this is reversed by allowing the stabilization of Hypoxia Inducible Factor-1 alpha (HIF-1α). We further show that on the long run CI-deficient tumors re-adapt to their inability to respond to hypoxia, concordantly with the persistence of human oncocytomas. We demonstrate that CI-deficient tumors survive and carry out angiogenesis, despite their inability to stabilize HIF-1α. Such adaptive response is mediated by tumor associated macrophages, whose blockage improves the effect of CI ablation. Additionally, the simultaneous pharmacological inhibition of CI function through metformin and macrophage infiltration through PLX-3397 impairs tumor growth in vivo in a synergistic manner, setting the basis for an efficient combinatorial adjuvant therapy in clinical trials.


Asunto(s)
Adenoma Oxifílico/tratamiento farmacológico , Adenoma Oxifílico/genética , Aminopiridinas/farmacología , Antineoplásicos/farmacología , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo I de Transporte de Electrón/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Metformina/farmacología , Pirroles/farmacología , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Drosophila , Femenino , Técnicas de Inactivación de Genes , Células HCT116 , Humanos , Macrófagos/inmunología , Ratones , Ratones Noqueados , Ratones Desnudos , NADH Deshidrogenasa/genética , Neovascularización Patológica/patología , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Cell Rep ; 13(11): 2456-2469, 2015 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-26670048

RESUMEN

During metastatic colonization, tumor cells must establish a favorable microenvironment or niche that will sustain their growth. However, both the temporal and molecular details of this process remain poorly understood. Here, we found that metastatic initiating cells (MICs) exhibit a high capacity for lung fibroblast activation as a result of Thrombospondin 2 (THBS2) expression. Importantly, inhibiting the mesenchymal phenotype of MICs by blocking the epithelial-to-mesenchymal transition (EMT)-associated kinase AXL reduces THBS2 secretion, niche-activating ability, and, consequently, metastatic competence. Subsequently, disseminated metastatic cells revert to an AXL-negative, more epithelial phenotype to proliferate and decrease the phosphorylation levels of TGF-ß-dependent SMAD2-3 in favor of BMP/SMAD1-5 signaling. Remarkably, newly activated fibroblasts promote this transition. In summary, our data reveal a crosstalk between cancer cells and their microenvironment whereby the EMT status initially triggers and then is regulated by niche activation during metastatic colonization.


Asunto(s)
Células Madre Neoplásicas/metabolismo , Trombospondinas/metabolismo , Animales , Benzocicloheptenos/toxicidad , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Antígeno CD24/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal , Femenino , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Ratones , Ratones Desnudos , Ratones Transgénicos , Metástasis de la Neoplasia , Células Madre Neoplásicas/citología , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Interferencia de ARN , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal , Proteínas Smad/genética , Proteínas Smad/metabolismo , Trombospondinas/antagonistas & inhibidores , Trombospondinas/genética , Factor de Crecimiento Transformador beta/metabolismo , Trasplante Heterólogo , Triazoles/toxicidad , Tirosina Quinasa del Receptor Axl
15.
Crit Rev Oncog ; 19(5): 349-61, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25404150

RESUMEN

Tumor metastasis, the cause of more than 90% of cancer cell mortality, is a multistep process by which tumor cells disseminate from their primary site via local invasion and intravasation into blood or lymphatic vessels and reach secondary distant sites, where they survive and reinitiate tumor growth. Activation of a developmental program called the epithelial-to-mesenchymal transition (EMT) has been shown to be a very efficient strategy adopted by epithelial cancer cells to promote local invasion and dissemination at distant organs. Remarkably, the activation of EMT programs in epithelial cells correlates with the appearance of stemness. This finding suggests that the EMT process also drives the initial cancer cell colonization at distant sites. However, recent studies support the concept that its reverse program, a mesenchymal-to-epithelial transition, is required for efficient metastatic colonization and that EMT is not necessarily associated with stemness. This review analyzes the conflicting experimental evidence linking epithelial plasticity to stemness in the light of an "EMT gradient model," according to which the outcome of EMT program activation in epithelial cells would be bimodal: coupled to stemness during initial activation, but when forced to reach an advanced mesenchymal status, it would become incompatible with stem cell abilities.


Asunto(s)
División Celular , Transición Epitelial-Mesenquimal , Metástasis de la Neoplasia , Neoplasias/patología , Humanos , Células Madre Neoplásicas/patología
16.
Stem Cell Reports ; 2(5): 707-20, 2014 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-24936456

RESUMEN

Cyclic activation of the Wnt/ß-catenin signaling pathway controls cell fusion-mediated somatic cell reprogramming. TCFs belong to a family of transcription factors that, in complex with ß-catenin, bind and transcriptionally regulate Wnt target genes. Here, we show that Wnt/ß-catenin signaling needs to be off during the early reprogramming phases of mouse embryonic fibroblasts (MEFs) into iPSCs. In MEFs undergoing reprogramming, senescence genes are repressed and mesenchymal-to-epithelial transition is favored. This is correlated with a repressive activity of TCF1, which contributes to the silencing of Wnt/ß-catenin signaling at the onset of reprogramming. In contrast, the Wnt pathway needs to be active in the late reprogramming phases to achieve successful reprogramming. In conclusion, continued activation or inhibition of the Wnt/ß-catenin signaling pathway is detrimental to the reprogramming of MEFs; instead, temporal perturbation of the pathway is essential for efficient reprogramming, and the "Wnt-off" state can be considered an early reprogramming marker.


Asunto(s)
Factor Nuclear 1-alfa del Hepatocito/metabolismo , Vía de Señalización Wnt , Animales , Antibióticos Antineoplásicos/farmacología , Línea Celular , Reprogramación Celular/efectos de los fármacos , Doxorrubicina/farmacología , Transición Epitelial-Mesenquimal , Factor Nuclear 1-alfa del Hepatocito/antagonistas & inhibidores , Factor Nuclear 1-alfa del Hepatocito/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
17.
Cell Cycle ; 11(1): 39-47, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22186782

RESUMEN

Wnt/ß-catenin canonical pathway has very important roles both in the regulation of self-renewal and lineage differentiation of pluripotent stem cells, as well as in the reprogramming of somatic cells. In the absence of Wnt stimulation, the target genes of the pathway are repressed or down-regulated. Conversely, when ß-catenin is stabilized there is an accumulation of cytoplasmic and nuclear ß-catenin that allows up-regulation of its target genes. However, the molecular mechanisms downstream of ß-catenin that regulate self-renewal of embryonic stem cells remain unclear. It has been proposed that ß-catenin acts through members of the T-cell factor (TCF) family. Recent studies have confirmed the role of Tcf3 (Tcf7l1) as a transcriptional repressor in embryonic stem cells, and they have also proposed a new mechanistic view as to how activation of the Wnt/ß-catenin pathway can overcome this repression.


Asunto(s)
Reprogramación Celular , Factores de Transcripción TCF/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Células Madre Embrionarias/metabolismo , Humanos , Factor 1 de Transcripción de Linfocitos T/metabolismo , Proteína 1 Similar al Factor de Transcripción 7/metabolismo , Regulación hacia Arriba , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo
18.
Proc Natl Acad Sci U S A ; 108(29): 11912-7, 2011 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-21730189

RESUMEN

The heterochromatin barrier must be overcome to generate induced pluripotent stem cells and cell fusion-mediated reprogrammed hybrids. Here, we show that the absence of T-cell factor 3 (Tcf3), a repressor of ß-catenin target genes, strikingly and rapidly enhances the efficiency of neural precursor cell (NPC) reprogramming. Remarkably, Tcf3(-/-) ES cells showed a genome-wide increase in AcH3 and decrease in H3K9me3 and can reprogram NPCs after fusion greatly. In addition, during reprogramming of NPCs into induced pluripotent stem cells, the silencing of Tcf3 increased AcH3 and decreased the number of H3K9me3-positive heterochromatin foci early and long before reactivation of the endogenous stem cell genes. In conclusion, our data suggest that Tcf3 functions as a repressor of the reprogramming potential of somatic cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Reprogramación Celular/fisiología , Epigénesis Genética/fisiología , Eliminación de Gen , Células Madre Pluripotentes Inducidas/fisiología , Neuronas/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Línea Celular , Reprogramación Celular/genética , Inmunoprecipitación de Cromatina , Epigénesis Genética/genética , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Vectores Genéticos/genética , Immunoblotting , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Retroviridae , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Pediatr Res ; 61(4): 410-4, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17515863

RESUMEN

Human milk stimulates intestinal development through the effects of various moieties. Lactoferrin (LF) is a glycoprotein of human milk whose concentration is highest in colostrum decreasing in mature milk. LF promotes enterocyte growth in intestinal cell lines. We tested the hypothesis that LF induces a distinct effect on enterocyte proliferation and differentiation, depending on its concentration. We examined the dose-related effects by human-native LF (N-LF) in Caco-2 (human colon adenocarcinoma) cells. At high concentrations, N-LF stimulated cell proliferation in immature Caco-2 cells, as judged by 3H-thymidine incorporation. In contrast, sucrase and lactase activities were increased at low but not high LF concentrations and their mRNA were also increased, indicating a transcriptional effect. Because iron binds specific LF sites, we compared the potency of N-LF and iron-saturated LF (I-LF) and found the native form more potent. Finally, we tested the effects by bovine LF (bLF) in the same system and found the latter more potent than the human isoform in inducing cell growth and lactase expression. These results suggest that LF directly induces enterocyte growth and proliferation, depending on its concentration, thereby regulating the earlyx postnatal intestinal development. bLF could be added to infant formula as a growth factor in selected intestinal diseases.


Asunto(s)
Diferenciación Celular/fisiología , Proliferación Celular , Enterocitos/citología , Enterocitos/metabolismo , Lactoferrina/fisiología , Animales , Células CACO-2 , Bovinos , Humanos , Lactoferrina/metabolismo , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...