Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancer Res ; 83(10): 1596-1610, 2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-36912618

RESUMEN

Cancer-associated fibroblasts (CAF) are a major cell type in the stroma of solid tumors and can exert both tumor-promoting and tumor-restraining functions. CAF heterogeneity is frequently observed in pancreatic ductal adenocarcinoma (PDAC), a tumor characterized by a dense and hypoxic stroma that features myofibroblastic CAFs (myCAF) and inflammatory CAFs (iCAF) that are thought to have opposing roles in tumor progression. While CAF heterogeneity can be driven in part by tumor cell-produced cytokines, other determinants shaping CAF identity and function are largely unknown. In vivo, we found that iCAFs displayed a hypoxic gene expression and biochemical profile and were enriched in hypoxic regions of PDAC tumors, while myCAFs were excluded from these regions. Hypoxia led fibroblasts to acquire an inflammatory gene expression signature and synergized with cancer cell-derived cytokines to promote an iCAF phenotype in a HIF1α-dependent fashion. Furthermore, HIF1α stabilization was sufficient to induce an iCAF phenotype in stromal cells introduced into PDAC organoid cocultures and to promote PDAC tumor growth. These findings indicate hypoxia-induced HIF1α as a regulator of CAF heterogeneity and promoter of tumor progression in PDAC. SIGNIFICANCE: Hypoxia in the tumor microenvironment of pancreatic cancer potentiates the cytokine-induced inflammatory CAF phenotype and promotes tumor growth. See related commentary by Fuentes and Taniguchi, p. 1560.


Asunto(s)
Fibroblastos Asociados al Cáncer , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Citocinas/metabolismo , Neoplasias Pancreáticas/patología , Carcinoma Ductal Pancreático/patología , Fibroblastos/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Fenotipo , Microambiente Tumoral , Neoplasias Pancreáticas
2.
bioRxiv ; 2023 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-36712084

RESUMEN

Preventing tumor cells from acquiring metastatic properties would significantly reduce cancer mortality. However, due to the complex nature of this process, it remains one of the most poorly understood and untreatable aspects of cancer. Ischemia and hypoxia in solid tumors are requisite in metastasis formation -- conditions that arise far from functional blood vessels and deep within tumor tissues. These secluded locations impede the observation of pre-metastatic tumor cells and their interactions with stromal cells, which are also critical in the initiation of this process. Thus, the initiation of metastasis has been incredibly difficult to model in the lab and to observe in vivo. We present an ex vivo model of the tumor microenvironment, called 3MIC, which overcomes these experimental challenges and enables the observation of ischemic tumor cells in their native 3D context with high spatial and temporal resolutions. The 3MIC recreates ischemic conditions in the tumor microenvironment and facilitates the co-culture of different cell types. Using live microscopy, we showed that ischemia, but not hypoxia alone, increases the motility and invasive properties of cells derived from primary tumors. These changes are phenotypic and can occur without clonal selection. We directly observed how interactions with stromal cells such as macrophages increased tumor invasion in conjunction with the effects of an ischemic microenvironment. Finally, we tested the effects of chemotherapy drugs under different metabolic microenvironments and found that ischemic tumor cells are more resistant to paclitaxel, possibly due to a metabolic resistance mechanism. Overall, the 3MIC is a cost-effective system that allows for the dissection of the complexity of the tumor microenvironment and direct observation of the emergence of metastasis, as well as the testing of treatments that may halt this process.

3.
Dis Model Mech ; 14(8)2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34407185

RESUMEN

There is an urgent need for accurate, scalable and cost-efficient models of the tumor microenvironment. Here, we detail how to fabricate and use the metabolic microenvironment chamber (MEMIC) - a 3D-printed ex vivo model of intratumoral heterogeneity. A major driver of the cellular and molecular diversity in tumors is accessibility to the blood stream. Whereas perivascular tumor cells have direct access to oxygen and nutrients, cells further from the vasculature must survive under progressively more ischemic environments. The MEMIC simulates this differential access to nutrients, allow co-culturing any number of cell types, and it is optimized for live imaging and other microscopy-based analyses. Owing to a modular design and full experimental control, the MEMIC provides insights into the tumor microenvironment that would be difficult to obtain via other methods. As proof of principle, we show that cells sense gradual changes in metabolite concentration leading to predictable molecular and cellular spatial patterns. We propose the MEMIC as a complement to standard in vitro and in vivo experiments, diversifying the tools available to accurately model, perturb and monitor the tumor microenvironment.


Asunto(s)
Neoplasias , Microambiente Tumoral , Técnicas de Cocultivo , Humanos , Neoplasias/patología
4.
Am Nat ; 194(3): 291-305, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31553215

RESUMEN

Predicting the evolution of expanding populations is critical to controlling biological threats such as invasive species and cancer metastasis. Expansion is primarily driven by reproduction and dispersal, but nature abounds with examples of evolution where organisms pay a reproductive cost to disperse faster. When does selection favor this "survival of the fastest"? We searched for a simple rule, motivated by evolution experiments where swarming bacteria evolved into a hyperswarmer mutant that disperses ∼100% faster but pays a growth cost of ∼10% to make many copies of its flagellum. We analyzed a two-species model based on the Fisher equation to explain this observation: the population expansion rate (v) results from an interplay of growth (r) and dispersal (D) and is independent of the carrying capacity: v=2(rD)1/2 . A mutant can take over the edge only if its expansion rate (v2) exceeds the expansion rate of the established species (v1); this simple condition ( v2>v1 ) determines the maximum cost in slower growth that a faster mutant can pay and still be able to take over. Numerical simulations and time-course experiments where we tracked evolution by imaging bacteria suggest that our findings are general: less favorable conditions delay but do not entirely prevent the success of the fastest. Thus, the expansion rate defines a traveling wave fitness, which could be combined with trade-offs to predict evolution of expanding populations.


Asunto(s)
Evolución Biológica , Modelos Teóricos , Pseudomonas aeruginosa/crecimiento & desarrollo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Mutación , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/fisiología , Selección Genética
5.
Nat Metab ; 1(7): 676-687, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31511848

RESUMEN

Most rapidly proliferating mammalian cells rely on the oxidation of exogenous glutamine to support cell proliferation. We previously found that culture of mouse embryonic stem cells (ESCs) in the presence of inhibitors against MEK and GSK3ß to maintain pluripotency reduces cellular reliance on glutamine for tricarboxylic acid (TCA) cycle anaplerosis, enabling ESCs to proliferate in the absence of exogenous glutamine. Here we show that reduced dependence on exogenous glutamine is a generalizable feature of pluripotent stem cells. Enhancing self-renewal, through either overexpression of pluripotency-associated transcription factors or altered signal transduction, decreases the utilization of glutamine-derived carbons in the TCA cycle. As a result, cells with the highest potential for self-renewal can be enriched by transient culture in glutamine-deficient media. During pluripotent cell culture or reprogramming to pluripotency, transient glutamine withdrawal selectively leads to the elimination of non-pluripotent cells. These data reveal that reduced dependence on glutamine anaplerosis is an inherent feature of self-renewing pluripotent stem cells and reveal a simple, non-invasive mechanism to select for mouse and human pluripotent stem cells within a heterogeneous population during both ESC passage and induced pluripotent cell reprogramming.


Asunto(s)
Glutamina/metabolismo , Células Madre Pluripotentes/metabolismo , Animales , Diferenciación Celular/fisiología , Reprogramación Celular , Humanos , Ratones , Células Madre Embrionarias de Ratones/metabolismo
6.
Nature ; 573(7775): 595-599, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31534224

RESUMEN

The tumour suppressor TP53 is mutated in the majority of human cancers, and in over 70% of pancreatic ductal adenocarcinoma (PDAC)1,2. Wild-type p53 accumulates in response to cellular stress, and regulates gene expression to alter cell fate and prevent tumour development2. Wild-type p53 is also known to modulate cellular metabolic pathways3, although p53-dependent metabolic alterations that constrain cancer progression remain poorly understood. Here we find that p53 remodels cancer-cell metabolism to enforce changes in chromatin and gene expression that favour a premalignant cell fate. Restoring p53 function in cancer cells derived from KRAS-mutant mouse models of PDAC leads to the accumulation of α-ketoglutarate (αKG, also known as 2-oxoglutarate), a metabolite that also serves as an obligate substrate for a subset of chromatin-modifying enzymes. p53 induces transcriptional programs that are characteristic of premalignant differentiation, and this effect can be partially recapitulated by the addition of cell-permeable αKG. Increased levels of the αKG-dependent chromatin modification 5-hydroxymethylcytosine (5hmC) accompany the tumour-cell differentiation that is triggered by p53, whereas decreased 5hmC characterizes the transition from premalignant to de-differentiated malignant lesions that is associated with mutations in Trp53. Enforcing the accumulation of αKG in p53-deficient PDAC cells through the inhibition of oxoglutarate dehydrogenase-an enzyme of the tricarboxylic acid cycle-specifically results in increased 5hmC, tumour-cell differentiation and decreased tumour-cell fitness. Conversely, increasing the intracellular levels of succinate (a competitive inhibitor of αKG-dependent dioxygenases) blunts p53-driven tumour suppression. These data suggest that αKG is an effector of p53-mediated tumour suppression, and that the accumulation of αKG in p53-deficient tumours can drive tumour-cell differentiation and antagonize malignant progression.


Asunto(s)
Carcinoma Ductal Pancreático , Diferenciación Celular/genética , Ácidos Cetoglutáricos/metabolismo , Neoplasias Pancreáticas , Proteína p53 Supresora de Tumor/metabolismo , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/fisiopatología , Línea Celular Tumoral , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Ensamble y Desensamble de Cromatina/genética , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Ácidos Cetoglutáricos/farmacología , Ratones , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/fisiopatología , Unión Proteica , Ácido Succínico/metabolismo , Activación Transcripcional
7.
Dis Model Mech ; 11(8)2018 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-29991530

RESUMEN

The extracellular space of solid tumors ranges from being well-nurtured to being completely ischemic and can serve as a source of intratumoral heterogeneity, determining the behavior and molecular profiles of malignant and stromal cells. Here, we discuss how the metabolic tumor microenvironment modulates the phenotypes of the immune cells that infiltrate tumors, with an emphasis on tumor-associated macrophages. These cells constitute a diverse population that has pro-tumoral and anti-inflammatory properties, and are likened to anti-inflammatory 'M2' macrophages. Recent findings show how different metabolic microenvironments specify an array of phenotypic changes in macrophages. In tumors, extracellular metabolite levels vary predictably according to proximity to the vasculature, and phenotypic changes in tumor-associated macrophages and in other immune cells are also predictable. We speculate that this 'metabolic axis' of macrophage polarization modulates - and is modulated by - the response to inflammatory cues, creating a wide variety of possible phenotypic states. Understanding how extracellular metabolites influence cell phenotypes allows us to predict how tumor-associated macrophages and other tumor cells might change, with the aim of harnessing this predictability for therapy. Overall, we describe an emerging picture in which chemokines, growth factors and the metabolic tumor microenvironment act together to determine the phenotypes of tumor-infiltrating immune cells.


Asunto(s)
Polaridad Celular , Macrófagos/inmunología , Macrófagos/patología , Animales , Humanos , Inflamación/metabolismo , Inflamación/patología , Redes y Vías Metabólicas , Microambiente Tumoral
8.
Proc Natl Acad Sci U S A ; 114(11): 2934-2939, 2017 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-28246332

RESUMEN

The genetic and phenotypic diversity of cells within tumors is a major obstacle for cancer treatment. Because of the stochastic nature of genetic alterations, this intratumoral heterogeneity is often viewed as chaotic. Here we show that the altered metabolism of cancer cells creates predictable gradients of extracellular metabolites that orchestrate the phenotypic diversity of cells in the tumor microenvironment. Combining experiments and mathematical modeling, we show that metabolites consumed and secreted within the tumor microenvironment induce tumor-associated macrophages (TAMs) to differentiate into distinct subpopulations according to local levels of ischemia and their position relative to the vasculature. TAMs integrate levels of hypoxia and lactate into progressive activation of MAPK signaling that induce predictable spatial patterns of gene expression, such as stripes of macrophages expressing arginase 1 (ARG1) and mannose receptor, C type 1 (MRC1). These phenotypic changes are functionally relevant as ischemic macrophages triggered tube-like morphogenesis in neighboring endothelial cells that could restore blood perfusion in nutrient-deprived regions where angiogenic resources are most needed. We propose that gradients of extracellular metabolites act as tumor morphogens that impose order within the microenvironment, much like signaling molecules convey positional information to organize embryonic tissues. Unearthing embryology-like processes in tumors may allow us to control organ-like tumor features such as tissue repair and revascularization and treat intratumoral heterogeneity.


Asunto(s)
Neoplasias/metabolismo , Neoplasias/patología , Microambiente Tumoral , Línea Celular Tumoral , Análisis por Conglomerados , Metabolismo Energético , Espacio Extracelular/metabolismo , Perfilación de la Expresión Génica , Humanos , Hipoxia/metabolismo , Ácido Láctico/metabolismo , Sistema de Señalización de MAP Quinasas , Macrófagos/metabolismo , Macrófagos/patología , Neoplasias/genética , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Oxígeno/metabolismo , Fenotipo , Transcriptoma , Microambiente Tumoral/genética
9.
Nat Chem Biol ; 13(5): 494-500, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28263965

RESUMEN

The metabolite 2-hydroxyglutarate (2HG) can be produced as either a D-R- or L-S- enantiomer, each of which inhibits α-ketoglutarate (αKG)-dependent enzymes involved in diverse biologic processes. Oncogenic mutations in isocitrate dehydrogenase (IDH) produce D-2HG, which causes a pathologic blockade in cell differentiation. On the other hand, oxygen limitation leads to accumulation of L-2HG, which can facilitate physiologic adaptation to hypoxic stress in both normal and malignant cells. Here we demonstrate that purified lactate dehydrogenase (LDH) and malate dehydrogenase (MDH) catalyze stereospecific production of L-2HG via 'promiscuous' reduction of the alternative substrate αKG. Acidic pH enhances production of L-2HG by promoting a protonated form of αKG that binds to a key residue in the substrate-binding pocket of LDHA. Acid-enhanced production of L-2HG leads to stabilization of hypoxia-inducible factor 1 alpha (HIF-1α) in normoxia. These findings offer insights into mechanisms whereby microenvironmental factors influence production of metabolites that alter cell fate and function.


Asunto(s)
Biocatálisis , Glutaratos/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Malato Deshidrogenasa/metabolismo , Cromatografía de Gases y Espectrometría de Masas , Humanos , Concentración de Iones de Hidrógeno , Ácidos Cetoglutáricos/química , Ácidos Cetoglutáricos/metabolismo , Estructura Molecular , Estereoisomerismo
10.
Development ; 143(12): 2194-205, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27122165

RESUMEN

Delamination of neural crest (NC) cells is a bona fide physiological model of epithelial-to-mesenchymal transition (EMT), a process that is influenced by Wnt/ß-catenin signalling. Using two in vivo models, we show that Wnt/ß-catenin signalling is transiently inhibited at the time of NC delamination. In attempting to define the mechanism underlying this inhibition, we found that the scaffold proteins Dact1 and Dact2, which are expressed in pre-migratory NC cells, are required for NC delamination in Xenopus and chick embryos, whereas they do not affect the motile properties of migratory NC cells. Dact1/2 inhibit Wnt/ß-catenin signalling upstream of the transcriptional activity of T cell factor (TCF), which is required for EMT to proceed. Dact1/2 regulate the subcellular distribution of ß-catenin, preventing ß-catenin from acting as a transcriptional co-activator to TCF, yet without affecting its stability. Together, these data identify a novel yet important regulatory element that inhibits ß-catenin signalling, which then affects NC delamination.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Cresta Neural/citología , Cresta Neural/metabolismo , Proteínas Wnt/metabolismo , Animales , Movimiento Celular , Núcleo Celular/metabolismo , Embrión de Pollo , Femenino , Células HEK293 , Humanos , Fracciones Subcelulares/metabolismo , Vía de Señalización Wnt , Xenopus laevis/embriología , Xenopus laevis/metabolismo , beta Catenina/metabolismo
11.
Dev Dyn ; 245(5): 580-9, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26933766

RESUMEN

BACKGROUND: During the initial stages zebrafish neurulation, neural plate cells undergo highly coordinated movements before they assemble into a multicellular solid neural rod. We have previously identified that the underlying mesoderm is critical to ensure such coordination and generate correct neural tube organization. However, how intertissue coordination is achieved in vivo during zebrafish neural tube morphogenesis is unknown. RESULTS: In this work, we use quantitative live imaging to study the coordinated movements of neural ectoderm and mesoderm during dorsal tissue convergence. We show the extracellular matrix components laminin and fibronectin that lie between mesoderm and neural plate act to couple the movements of neural plate and mesoderm during early stages of neurulation and to maintain the close apposition of these two tissues. CONCLUSIONS: Our study highlights the importance of the extracellular matrix proteins laminin and libronectin in coupling the movements and spatial proximity of mesoderm and neuroectoderm during the morphogenetic movements of neurulation. Developmental Dynamics 245:580-589, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Matriz Extracelular/fisiología , Mesodermo/metabolismo , Placa Neural/metabolismo , Neurulación , Animales , Embrión no Mamífero , Fibronectinas/fisiología , Laminina/fisiología , Morfogénesis , Tubo Neural , Pez Cebra
12.
PLoS One ; 9(9): e104969, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25181349

RESUMEN

Collective cell migration is a fundamental process, occurring during embryogenesis and cancer metastasis. Neural crest cells exhibit such coordinated migration, where aberrant motion can lead to fatality or dysfunction of the embryo. Migration involves at least two complementary mechanisms: contact inhibition of locomotion (a repulsive interaction corresponding to a directional change of migration upon contact with a reciprocating cell), and co-attraction (a mutual chemoattraction mechanism). Here, we develop and employ a parameterized discrete element model of neural crest cells, to investigate how these mechanisms contribute to long-range directional migration during development. Motion is characterized using a coherence parameter and the time taken to reach, collectively, a target location. The simulated cell group is shown to switch from a diffusive to a persistent state as the response-rate to co-attraction is increased. Furthermore, the model predicts that when co-attraction is inhibited, neural crest cells can migrate into restrictive regions. Indeed, inhibition of co-attraction in vivo and in vitro leads to cell invasion into restrictive areas, confirming the prediction of the model. This suggests that the interplay between the complementary mechanisms may contribute to guidance of the neural crest. We conclude that directional migration is a system property and does not require action of external chemoattractants.


Asunto(s)
Comunicación Celular , Movimiento Celular , Animales , Calibración , Inhibición de Contacto , Modelos Biológicos , Factores de Tiempo , Xenopus
13.
Neural Dev ; 9: 9, 2014 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-24755297

RESUMEN

BACKGROUND: Morphogenesis of the zebrafish neural tube requires the coordinated movement of many cells in both time and space. A good example of this is the movement of the cells in the zebrafish neural plate as they converge towards the dorsal midline before internalizing to form a neural keel. How these cells are regulated to ensure that they move together as a coherent tissue is unknown. Previous work in other systems has suggested that the underlying mesoderm may play a role in this process but this has not been shown directly in vivo. RESULTS: Here we analyze the roles of subjacent mesoderm in the coordination of neural cell movements during convergence of the zebrafish neural plate and neural keel formation. Live imaging demonstrates that the normal highly coordinated movements of neural plate cells are lost in the absence of underlying mesoderm and the movements of internalization and neural tube formation are severely disrupted. Despite this, neuroepithelial polarity develops in the abnormal neural primordium but the resulting tissue architecture is very disorganized. CONCLUSIONS: We show that the movements of cells in the zebrafish neural plate are highly coordinated during the convergence and internalization movements of neurulation. Our results demonstrate that the underlying mesoderm is required for these coordinated cell movements in the zebrafish neural plate in vivo.


Asunto(s)
Movimiento Celular , Mesodermo/embriología , Placa Neural/embriología , Tubo Neural/embriología , Pez Cebra/embriología , Animales , Tipificación del Cuerpo , Ligandos de Señalización Nodal/metabolismo
14.
Soft Matter ; 10(14): 2405-13, 2014 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-24622509

RESUMEN

Pseudomonas aeruginosa is a monoflagellated bacterium that can use its single polar flagellum to swim through liquids and move collectively over semisolid surfaces, a behavior called swarming. Previous studies have shown that experimental evolution in swarming colonies leads to the selection of hyperswarming bacteria with multiple flagella. Here we show that the advantage of such hyperswarmer mutants cannot be explained simply by an increase in the raw swimming speed of individual bacteria in liquids. Cell tracking of time-lapse microscopy to quantify single-cell swimming patterns reveals that both wild-type and hyperswarmers alternate between forward and backward runs, rather than doing the run-and-tumble characteristic of enteric bacteria such as E. coli. High-throughput measurement of swimming speeds reveals that hyperswarmers do not swim faster than wild-type in liquid. Wild-type reverses swimming direction in sharp turns without a significant impact on its speed, whereas multiflagellated hyperswarmers tend to alternate fast and slow runs and have wider turning angles. Nonetheless, macroscopic measurement of swimming and swarming speed in colonies shows that hyperswarmers expand faster than wild-type on surfaces and through soft agar matrices. A mathematical model explains how wider turning angles lead to faster spreading when swimming through agar. Our study describes for the first time the swimming patterns in multiflagellated P. aeruginosa mutants and reveals that collective and individual motility in bacteria are not necessarily correlated. Understanding bacterial adaptations to surface motility, such as hyperswarming, requires a collective behavior approach.


Asunto(s)
Adaptación Fisiológica , Adhesión Bacteriana , Pseudomonas aeruginosa/fisiología , Modelos Biológicos , Mutación , Pseudomonas aeruginosa/genética , Percepción de Quorum
15.
Proc Natl Acad Sci U S A ; 110(48): 19402-7, 2013 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-24218566

RESUMEN

Drastic metabolic alterations, such as the Warburg effect, are found in most if not all types of malignant tumors. Emerging evidence shows that cancer cells benefit from these alterations, but little is known about how they affect noncancerous stromal cells within the tumor microenvironment. Here we show that cancer cells are better adapted to metabolic changes in the microenvironment, leading to the emergence of spatial structure. A clear example of tumor spatial structure is the localization of tumor-associated macrophages (TAMs), one of the most common stromal cell types found in tumors. TAMs are enriched in well-perfused areas, such as perivascular and cortical regions, where they are known to potentiate tumor growth and invasion. However, the mechanisms of TAM localization are not completely understood. Computational modeling predicts that gradients--of nutrients, gases, and metabolic by-products such as lactate--emerge due to altered cell metabolism within poorly perfused tumors, creating ischemic regions of the tumor microenvironment where TAMs struggle to survive. We tested our modeling prediction in a coculture system that mimics the tumor microenvironment. Using this experimental approach, we showed that a combination of metabolite gradients and differential sensitivity to lactic acid is sufficient for the emergence of macrophage localization patterns in vitro. This suggests that cancer metabolic changes create a microenvironment where tumor cells thrive over other cells. Understanding differences in tumor-stroma sensitivity to these alterations may open therapeutic avenues against cancer.


Asunto(s)
Glucólisis/fisiología , Macrófagos/fisiología , Modelos Biológicos , Neoplasias/metabolismo , Microambiente Tumoral/fisiología , Línea Celular Tumoral , Técnica del Anticuerpo Fluorescente , Humanos , Concentración de Iones de Hidrógeno , Procesamiento de Imagen Asistido por Computador , Macrófagos/citología , Microscopía Fluorescente , Estadísticas no Paramétricas
16.
Interface Focus ; 3(4): 20130017, 2013 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-24511381

RESUMEN

Cancer cell collective migration is a complex behaviour leading to the invasion of cancer cells into surrounding tissue, often with the aid of stromal cells in the microenvironment, such as macrophages or fibroblasts. Although tumour-tumour and tumour-stromal intercellular signalling have been shown to contribute to cancer cell migration, we lack a fundamental theoretical understanding of how aggressive invasion emerges from the synergy between these mechanisms. We use a computational self-propelled particle model to simulate intercellular interactions between co-migrating tumour and stromal cells and study the emergence of collective movement. We find that tumour-stromal interaction increases the cohesion and persistence of migrating mixed tumour-stromal cell clusters in a noisy and unbounded environment, leading to increased cell cluster size and distance migrated by cancer cells. Although environmental constraints, such as vasculature or extracellular matrix, influence cancer migration in vivo, our model shows that cell-cell interactions are sufficient to generate cohesive and persistent movement. From our results, we conclude that inhibition of tumour-stromal intercellular signalling may present a viable therapeutic target for disrupting collective cancer cell migration.

18.
Dev Cell ; 21(6): 1026-37, 2011 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-22118769

RESUMEN

Collective cell migration is a mode of movement crucial for morphogenesis and cancer metastasis. However, little is known about how migratory cells coordinate collectively. Here we show that mutual cell-cell attraction (named here coattraction) is required to maintain cohesive clusters of migrating mesenchymal cells. Coattraction can counterbalance the natural tendency of cells to disperse via mechanisms such as contact inhibition and epithelial-to-mesenchymal transition. Neural crest cells are coattracted via the complement fragment C3a and its receptor C3aR, revealing an unexpected role of complement proteins in early vertebrate development. Loss of coattraction disrupts collective and coordinated movements of these cells. We propose that coattraction and contact inhibition act in concert to allow cell collectives to self-organize and respond efficiently to external signals, such as chemoattractants and repellents.


Asunto(s)
Comunicación Celular/fisiología , Movimiento Celular/fisiología , Complemento C3a/fisiología , Animales , Adhesión Celular/fisiología , Factores Quimiotácticos/genética , Factores Quimiotácticos/fisiología , Complemento C3a/genética , Transición Epitelial-Mesenquimal/fisiología , Modelos Neurológicos , Datos de Secuencia Molecular , Células Madre Multipotentes/fisiología , Cresta Neural/citología , Cresta Neural/embriología , Células-Madre Neurales/fisiología , Receptores de Complemento/genética , Receptores de Complemento/fisiología , Proteínas de Xenopus/genética , Proteínas de Xenopus/fisiología , Xenopus laevis/embriología , Xenopus laevis/genética , Xenopus laevis/fisiología , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/fisiología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/fisiología
19.
Trends Cell Biol ; 20(6): 319-28, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20399659

RESUMEN

Contact inhibition of locomotion (CIL) is the process by which cells in vitro change their direction of migration upon contact with another cell. Here, we revisit the concept that CIL plays a central role in the migration of single cells and in collective migration, during both health and disease. Importantly, malignant cells exhibit a diminished CIL behaviour which allows them to invade healthy tissues. Accumulating evidence indicates that CIL occurs in vivo and that regulation of small Rho GTPases is important in the collapse of cell protrusions upon cell contact, the first step of CIL. Finally, we propose possible cell surface proteins that could be involved in the initial contact that regulates Rho GTPases during CIL.


Asunto(s)
Movimiento Celular , Inhibición de Contacto , Animales , Polaridad Celular , Humanos , Proteínas de la Membrana/metabolismo , Neoplasias/patología
20.
Nature ; 456(7224): 957-61, 2008 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-19078960

RESUMEN

Contact inhibition of locomotion was discovered by Abercrombie more than 50 years ago and describes the behaviour of fibroblast cells confronting each other in vitro, where they retract their protrusions and change direction on contact. Its failure was suggested to contribute to malignant invasion. However, the molecular basis of contact inhibition of locomotion and whether it also occurs in vivo are still unknown. Here we show that neural crest cells, a highly migratory and multipotent embryonic cell population, whose behaviour has been likened to malignant invasion, demonstrate contact inhibition of locomotion both in vivo and in vitro, and that this accounts for their directional migration. When two migrating neural crest cells meet, they stop, collapse their protrusions and change direction. In contrast, when a neural crest cell meets another cell type, it fails to display contact inhibition of locomotion; instead, it invades the other tissue, in the same manner as metastatic cancer cells. We show that inhibition of non-canonical Wnt signalling abolishes both contact inhibition of locomotion and the directionality of neural crest migration. Wnt-signalling members localize at the site of cell contact, leading to activation of RhoA in this region. These results provide the first example of contact inhibition of locomotion in vivo, provide an explanation for coherent directional migration of groups of cells and establish a previously unknown role for non-canonical Wnt signalling.


Asunto(s)
Movimiento Celular , Inhibición de Contacto , Cresta Neural/citología , Animales , Comunicación Celular , Polaridad Celular , Embrión no Mamífero/citología , Transducción de Señal , Proteínas Wnt/metabolismo , Xenopus/embriología , Pez Cebra/embriología , Proteína de Unión al GTP rhoA/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...