Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Cancers (Basel) ; 15(6)2023 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-36980785

RESUMEN

Cancer cells rely on the tumor microenvironment (TME), a composite of non-malignant cells, and extracellular matrix (ECM), for survival, growth, and metastasis. The ECM contributes to the biomechanical properties of the surrounding tissue, in addition to providing signals for tissue development. Cancer-associated fibroblasts (CAFs) are stromal cells in the TME that are integral to cancer progression. Subtypes of CAFs across a variety of cancers have been revealed, and each play a different role in cancer progression or suppression. CAFs secrete signaling molecules and remodel the surrounding ECM by depositing its constituents as well as degrading enzymes. In cancer, a remodeled ECM can lead to tumor-promoting effects. Not only does the remodeled ECM promote growth and allow for easier metastasis, but it can also modulate the immune system. A better understanding of how CAFs remodel the ECM will likely yield novel therapeutic targets. In this review, we summarize the key factors secreted by CAFs that facilitate tumor progression, ECM remodeling, and immune suppression.

2.
Transl Lung Cancer Res ; 11(6): 991-1008, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35832452

RESUMEN

Background: The microanatomical steps of malignant pleural mesothelioma (MPM) vascularization and the resistance mechanisms to anti-angiogenic drugs in MPM are unclear. Methods: We investigated the vascularization of intrapleurally implanted human P31 and SPC111 MPM cells. We also assessed MPM cell's motility, invasion and interaction with endothelial cells in vitro. Results: P31 cells exhibited significantly higher two-dimensional (2D) motility and three-dimensional (3D) invasion than SPC111 cells in vitro. In co-cultures of MPM and endothelial cells, P31 spheroids permitted endothelial sprouting (ES) with minimal spatial distortion, whereas SPC111 spheroids repealed endothelial sprouts. Both MPM lines induced the early onset of submesothelial microvascular plexuses covering large pleural areas including regions distant from tumor colonies. The development of these microvascular networks occurred due to both intussusceptive angiogenesis (IA) and ES and was accelerated by vascular endothelial growth factor A (VEGF-A)-overexpression. Notably, SPC111 colonies showed different behavior to P31 cells. P31 nodules incorporated tumor-induced capillary plexuses from the earliest stages of tumor formation. P31 cells deposited a collagenous matrix of human origin which provided "space" for further intratumoral angiogenesis. In contrast, SPC111 colonies pushed the capillary plexuses away and thus remained avascular for weeks. The key event in SPC111 vascularization was the development of a desmoplastic matrix of mouse origin. Continuously invaded by SPC111 cells, this matrix transformed into intratumoral connective tissue trunks, providing a route for ES from the diaphragm. Conclusions: Here, we report two distinct growth patterns of orthotopically implanted human MPM xenografts. In the invasive pattern, MPM cells invade and thus co-opt peritumoral capillary plexuses. In the pushing/desmoplastic pattern, MPM cells induce a desmoplastic response within the underlying tissue which allows the ingrowth of a nutritive vasculature from the pleura.

3.
Front Cell Dev Biol ; 10: 852812, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35392170

RESUMEN

Malignant pleural mesothelioma (MPM) is a rare type of cancer with a grim prognosis. So far, no targetable oncogenic mutation was identified in MPM and biomarkers with predictive value toward drug sensitivity or resistance are also lacking. Nintedanib (BIBF1120) is a small-molecule tyrosine kinase inhibitor that showed promising efficacy preclinically and in phase II trial in MPM as an angiogenesis inhibitor combined with chemotherapy. However, the extended phase III trial failed. In this study, we investigated the effect of nintedanib on one of its targets, the SRC kinase, in two commercial and six novel MPM cell lines. Surprisingly, nintedanib treatment did not inhibit SRC activation in MPM cells and even increased phosphorylation of SRC in several cell lines. Combination treatment with the SRC inhibitor dasatinib could reverse this effect in all cell lines, however, the cellular response was dependent on the drug sensitivity of the cells. In 2 cell lines, with high sensitivity to both nintedanib and dasatinib, the drug combination had no synergistic effect but cell death was initiated. In 2 cell lines insensitive to nintedanib combination treatment reduced cell viability synergisticaly without cell death. In contrast, in these cells both treatments increased the autophagic flux assessed by degradation of the autophagy substrate p62 and increased presence of LC3B-II, increased number of GFP-LC3 puncta and decreased readings of the HiBiT-LC3 reporter. Additionaly, autophagy was synergistically promoted by the combined treatment. At the transcriptional level, analysis of lysosomal biogenesis regulator Transcription Factor EB (TFEB) showed that in all cell lines treated with nintedanib and to a lesser extent, with dasatinib, it became dephosphorylated and accumulated in the nucleus. Interestingly, the expression of certain known TFEB target genes implicated in autophagy or lysosomal biogenesis were significantly modified only in 1 cell line. Finally, we showed that autophagy induction in our MPM cell lines panel by nintedanib and dasatinib is independent of the AKT/mTOR and the ERK pathways. Our study reveals that autophagy can serve as a cytoprotective mechanism following nintedanib or dasatinib treatments in MPM cells.

4.
Sci Rep ; 11(1): 5798, 2021 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-33707612

RESUMEN

Apelin, a ligand of the APJ receptor, is overexpressed in several human cancers and plays an important role in tumor angiogenesis and growth in various experimental systems. We investigated the role of apelin signaling in the malignant behavior of cutaneous melanoma. Murine B16 and human A375 melanoma cell lines were stably transfected with apelin encoding or control vectors. Apelin overexpression significantly increased melanoma cell migration and invasion in vitro, but it had no impact on its proliferation. In our in vivo experiments, apelin significantly increased the number and size of lung metastases of murine melanoma cells. Melanoma cell proliferation rates and lymph and blood microvessel densities were significantly higher in the apelin-overexpressing pulmonary metastases. APJ inhibition by the competitive APJ antagonist MM54 significantly attenuated the in vivo pro-tumorigenic effects of apelin. Additionally, we detected significantly elevated circulating apelin and VEGF levels in patients with melanoma compared to healthy controls. Our results show that apelin promotes blood and lymphatic vascularization and the growth of pulmonary metastases of skin melanoma. Further studies are warranted to validate apelin signaling as a new potential therapeutic target in this malignancy.


Asunto(s)
Apelina/efectos adversos , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/secundario , Linfangiogénesis , Melanoma Experimental/patología , Neovascularización Patológica/patología , Animales , Estudios de Casos y Controles , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Humanos , Neoplasias Pulmonares/sangre , Masculino , Melanoma Experimental/sangre , Ratones , Persona de Mediana Edad , Invasividad Neoplásica , Neovascularización Patológica/sangre , Factor A de Crecimiento Endotelial Vascular/sangre
5.
Sci Rep ; 10(1): 20114, 2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-33208866

RESUMEN

Malignant pleural mesothelioma (MPM) has an overall poor prognosis and unsatisfactory treatment options. MPM nodules, protruding into the pleural cavity may have growth and spreading dynamics distinct that of other solid tumors. We demonstrate that multicellular aggregates can develop spontaneously in the majority of tested MPM cell lines when cultured at high cell density. Surprisingly, the nodule-like aggregates do not arise by excessive local cell proliferation, but by myosin II-driven cell contractility. Prominent actin cables, spanning several cells, are abundant both in cultured aggregates and in MPM surgical specimens. We propose a computational model for in vitro MPM nodule development. Such a self-tensioned Maxwell fluid exhibits a pattern-forming instability that was studied by analytical tools and computer simulations. Altogether, our findings may underline a rational for targeting the actomyosin system in MPM.


Asunto(s)
Mesotelioma Maligno/patología , Actinas/metabolismo , Amidas/farmacología , Animales , Recuento de Células , Línea Celular Tumoral , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Masculino , Mesotelioma Maligno/metabolismo , Ratones SCID , Miosinas/metabolismo , Piridinas/farmacología , Procesos Estocásticos , Imagen de Lapso de Tiempo , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Sci Rep ; 10(1): 8128, 2020 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-32424155

RESUMEN

The collective motion of cell monolayers within a tissue is a fundamental biological process that occurs during tissue formation, wound healing, cancerous invasion, and viral infection. Experiments have shown that at the onset of migration, the motility is self-generated as a polarisation wave starting from the leading edge of the monolayer and progressively propagates into the bulk. However, it is unclear how the propagation of this motility wave is influenced by cellular properties. Here, we investigate this question using a computational model based on the Potts model coupled to the dynamics of intracellular polarisation. The model captures the propagation of the polarisation wave and suggests that the cells cortex can regulate the migration modes: strongly contractile cells may depolarise the monolayer, whereas less contractile cells can form swirling movement. Cortical contractility is further found to limit the cells motility, which (i) decelerates the wave speed and the leading edge progression, and (ii) destabilises the leading edge. Together, our model describes how different mechanical properties of cells can contribute to the regulation of collective cell migration.


Asunto(s)
Movimiento Celular , Células/química , Células/citología , Células Epiteliales/química , Células Epiteliales/citología , Fenómenos Biomecánicos , Polaridad Celular , Forma de la Célula , Simulación por Computador , Humanos , Modelos Biológicos
7.
Cells ; 8(11)2019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31671862

RESUMEN

Epithelial to mesenchymal transition (EMT) is a multipurpose process involved in wound healing, development, and certain pathological processes, such as metastasis formation. The Tks4 scaffold protein has been implicated in cancer progression; however, its role in oncogenesis is not well defined. In this study, the function of Tks4 was investigated in HCT116 colon cancer cells by knocking the protein out using the CRISPR/Cas9 system. Surprisingly, the absence of Tks4 induced significant changes in cell morphology, motility, adhesion and expression, and localization of E-cadherin, which are all considered as hallmarks of EMT. In agreement with these findings, the marked appearance of fibronectin, a marker of the mesenchymal phenotype, was also observed in Tks4-KO cells. Analysis of the expression of well-known EMT transcription factors revealed that Snail2 was strongly overexpressed in cells lacking Tks4. Tks4-KO cells showed increased motility and decreased cell-cell attachment. Collagen matrix invasion assays demonstrated the abundance of invasive solitary cells. Finally, the reintroduction of Tks4 protein in the Tks4-KO cells restored the expression levels of relevant key transcription factors, suggesting that the Tks4 scaffold protein has a specific and novel role in EMT regulation and cancer progression.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Neoplasias del Colon/genética , Transición Epitelial-Mesenquimal/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Movimiento Celular/genética , Transformación Celular Neoplásica/genética , Neoplasias del Colon/patología , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Células HCT116 , Humanos , Invasividad Neoplásica , Transducción de Señal/genética
8.
PLoS Comput Biol ; 15(10): e1007431, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31652274

RESUMEN

Non-muscle myosin II (NMII)-induced multicellular contractility is essential for development, maintenance and remodeling of tissue morphologies. Dysregulation of the cytoskeleton can lead to birth defects or enable cancer progression. We demonstrate that the Matrigel patterning assay, widely used to characterize endothelial cells, is a highly sensitive tool to evaluate cell contractility within a soft extracellular matrix (ECM) environment. We propose a computational model to explore how cell-exerted contractile forces can tear up the cell-Matrigel composite material and gradually remodel it into a network structure. We identify measures that are characteristic for cellular contractility and can be obtained from image analysis of the recorded patterning process. The assay was calibrated by inhibition of NMII activity in A431 epithelial carcinoma cells either directly with blebbistatin or indirectly with Y27632 Rho kinase inhibitor. Using Matrigel patterning as a bioassay, we provide the first functional demonstration that overexpression of S100A4, a calcium-binding protein that is frequently overexpressed in metastatic tumors and inhibits NMIIA activity by inducing filament disassembly, effectively reduces cell contractility.


Asunto(s)
Bioensayo/métodos , Colágeno/fisiología , Proteínas Contráctiles/fisiología , Laminina/fisiología , Proteoglicanos/fisiología , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animales , Línea Celular Tumoral , Simulación por Computador , Citoesqueleto/metabolismo , Combinación de Medicamentos , Células Epiteliales/fisiología , Humanos , Ratones , Microtúbulos/metabolismo , Miosina Tipo IIA no Muscular/metabolismo , Proteína de Unión al Calcio S100A4/metabolismo
9.
Phys Rev E ; 100(3-1): 032403, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31640045

RESUMEN

Collective cell migration underlies morphogenesis, tissue regeneration, and cancer progression. How the biomechanical coupling between epithelial cells triggers and coordinates the collective migration is an open question. Here, we develop a one-dimensional model for an epithelial monolayer which predicts that after the onset of migration at an open boundary, cells in the bulk of the epithelium are gradually recruited into outward-directed motility, exhibiting traveling-wave-like behavior. We find an exact formula for the speed of this motility wave proportional to the square root of the cells' contractility, which accounts for cortex tension and adhesion between adjacent cells.


Asunto(s)
Movimiento Celular , Modelos Biológicos , Células Epiteliales/citología
10.
Sci Rep ; 9(1): 13188, 2019 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-31515494

RESUMEN

Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have been developed for cardiac cell transplantation studies more than a decade ago. In order to establish the hiPSC-CM-based platform as an autologous source for cardiac repair and drug toxicity, it is vital to understand the functionality of cardiomyocytes. Therefore, the goal of this study was to assess functional physiology, ultrastructural morphology, gene expression, and microRNA (miRNA) profiling at Wk-1, Wk-2 & Wk-4 in hiPSC-CMs in vitro. Functional assessment of hiPSC-CMs was determined by multielectrode array (MEA), Ca2+ cycling and particle image velocimetry (PIV). Results demonstrated that Wk-4 cardiomyocytes showed enhanced synchronization and maturation as compared to Wk-1 & Wk-2. Furthermore, ultrastructural morphology of Wk-4 cardiomyocytes closely mimicked the non-failing (NF) adult human heart. Additionally, modulation of cardiac genes, cell cycle genes, and pluripotency markers were analyzed by real-time PCR and compared with NF human heart. Increasing expression of fatty acid oxidation enzymes at Wk-4 supported the switching to lipid metabolism. Differential regulation of 12 miRNAs was observed in Wk-1 vs Wk-4 cardiomyocytes. Overall, this study demonstrated that Wk-4 hiPSC-CMs showed improved functional, metabolic and ultrastructural maturation, which could play a crucial role in optimizing timing for cell transplantation studies and drug screening.


Asunto(s)
Diferenciación Celular , Perfilación de la Expresión Génica , Células Madre Pluripotentes Inducidas/metabolismo , MicroARNs/biosíntesis , Miocitos Cardíacos/metabolismo , Línea Celular , Humanos , Células Madre Pluripotentes Inducidas/citología , MicroARNs/genética , Miocitos Cardíacos/citología
11.
Stem Cells ; 37(7): 910-923, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31087611

RESUMEN

Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) exhibit a fetal phenotype that limits in vitro and therapeutic applications. Strategies to promote cardiomyocyte maturation have focused interventions on differentiated hPSC-CMs, but this study tests priming of early cardiac progenitor cells (CPCs) with polyinosinic-polycytidylic acid (pIC) to accelerate cardiomyocyte maturation. CPCs were differentiated from hPSCs using a monolayer differentiation protocol with defined small molecule Wnt temporal modulation, and pIC was added during the formation of early CPCs. pIC priming did not alter the expression of cell surface markers for CPCs (>80% KDR+/PDGFRα+), expression of common cardiac transcription factors, or final purity of differentiated hPSC-CMs (∼90%). However, CPC differentiation in basal medium revealed that pIC priming resulted in hPSC-CMs with enhanced maturity manifested by increased cell size, greater contractility, faster electrical upstrokes, increased oxidative metabolism, and more mature sarcomeric structure and composition. To investigate the mechanisms of CPC priming, RNAseq revealed that cardiac progenitor-stage pIC modulated early Notch signaling and cardiomyogenic transcriptional programs. Chromatin immunoprecipitation of CPCs showed that pIC treatment increased deposition of the H3K9ac activating epigenetic mark at core promoters of cardiac myofilament genes and the Notch ligand, JAG1. Inhibition of Notch signaling blocked the effects of pIC on differentiation and cardiomyocyte maturation. Furthermore, primed CPCs showed more robust formation of hPSC-CMs grafts when transplanted to the NSGW mouse kidney capsule. Overall, epigenetic modulation of CPCs with pIC accelerates cardiomyocyte maturation enabling basic research applications and potential therapeutic uses. Stem Cells 2019;37:910-923.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Epigénesis Genética , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Poli I-C/farmacología , Receptores Notch/genética , Animales , Tamaño de la Célula , Histonas/genética , Histonas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/trasplante , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Riñón , Ratones , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Fosforilación Oxidativa , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores Notch/metabolismo , Sarcómeros/metabolismo , Análisis de Secuencia de ARN , Transducción de Señal , Trasplante de Células Madre/métodos , Trasplante Heterotópico , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
12.
J Theor Biol ; 456: 261-278, 2018 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-30086288

RESUMEN

Vascular patterning is a key process during development and disease. The diffusive decoy receptor sVEGFR1 (sFlt1) is a known regulator of endothelial cell behavior, yet the mechanism by which it controls vascular structure is little understood. We propose computational models to shed light on how vascular patterning is guided by self-organized gradients of the VEGF/sVEGFR1 factors. We demonstrate that a diffusive inhibitor can generate structures with a dense branching morphology in models where the activator elicits directed growth. Inadequate presence of the inhibitor leads to compact growth, while excessive production of the inhibitor blocks expansion and stabilizes existing structures. Model predictions were compared with time-resolved experimental data obtained from endothelial sprout kinetics in fibrin gels. In the presence of inhibitory antibodies against VEGFR1 vascular sprout density increases while the speed of sprout expansion remains unchanged. Thus, the rate of secretion and stability of extracellular sVEGFR1 can modulate vascular sprout density.


Asunto(s)
Modelos Cardiovasculares , Neovascularización Patológica/metabolismo , Neovascularización Fisiológica/fisiología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/fisiología , Algoritmos , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Neovascularización Patológica/patología , Transducción de Señal/fisiología , Esferoides Celulares/fisiología
13.
Clin Cancer Res ; 24(15): 3729-3740, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29724868

RESUMEN

Purpose: Malignant pleural mesothelioma (MPM) is an aggressive thoracic tumor type with limited treatment options and poor prognosis. The angiokinase inhibitor nintedanib has shown promising activity in the LUME-Meso phase II MPM trial and thus is currently being evaluated in the confirmatory LUME-Meso phase III trial. However, the anti-MPM potential of nintedanib has not been studied in the preclinical setting.Experimental Design: We have examined the antineoplastic activity of nintedanib in various in vitro and in vivo models of human MPM.Results: Nintedanib's target receptors were (co)expressed in all the 20 investigated human MPM cell lines. Nintedanib inhibited MPM cell growth in both short- and long-term viability assays. Reduced MPM cell proliferation and migration and the inhibition of Erk1/2 phosphorylation were also observed upon nintedanib treatment in vitro Additive effects on cell viability were detected when nintedanib was combined with cisplatin, a drug routinely used for systemic MPM therapy. In an orthotopic mouse model of human MPM, survival of animals receiving nintedanib per os showed a favorable trend, but no significant benefit. Nintedanib significantly reduced tumor burden and vascularization and prolonged the survival of mice when it was administered intraperitoneally. Importantly, unlike bevacizumab, nintedanib demonstrated significant in vivo antivascular and antitumor potential independently of baseline VEGF-A levels.Conclusions: Nintedanib exerts significant antitumor activity in MPM both in vitro and in vivo These data provide preclinical support for the concept of LUME-Meso trials evaluating nintedanib in patients with unresectable MPM. Clin Cancer Res; 24(15); 3729-40. ©2018 AACR.


Asunto(s)
Indoles/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Mesotelioma/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Neoplasias Pleurales/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cisplatino/farmacología , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Mesotelioma/genética , Mesotelioma/patología , Mesotelioma Maligno , Ratones , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Fosforilación/efectos de los fármacos , Neoplasias Pleurales/genética , Neoplasias Pleurales/patología , Ensayos Antitumor por Modelo de Xenoinjerto
14.
PLoS Comput Biol ; 13(11): e1005818, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29149169

RESUMEN

Resection of the bulk of a tumour often cannot eliminate all cancer cells, due to their infiltration into the surrounding healthy tissue. This may lead to recurrence of the tumour at a later time. We use a reaction-diffusion equation based model of tumour growth to investigate how the invasion front is delayed by resection, and how this depends on the density and behaviour of the remaining cancer cells. We show that the delay time is highly sensitive to qualitative details of the proliferation dynamics of the cancer cell population. The typically assumed logistic type proliferation leads to unrealistic results, predicting immediate recurrence. We find that in glioblastoma cell cultures the cell proliferation rate is an increasing function of the density at small cell densities. Our analysis suggests that cooperative behaviour of cancer cells, analogous to the Allee effect in ecology, can play a critical role in determining the time until tumour recurrence.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Biología Computacional/métodos , Glioblastoma/metabolismo , Modelos Biológicos , Recurrencia Local de Neoplasia/metabolismo , Algoritmos , Neoplasias Encefálicas/cirugía , Proliferación Celular , Difusión , Glioblastoma/cirugía , Humanos
15.
Sci Rep ; 7(1): 7358, 2017 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-28779157

RESUMEN

Short and long distance cell dispersal can have a marked effect on tumor structure, high cellular motility could lead to faster cell mixing and lower observable intratumor heterogeneity. Here we evaluated a model for cell mixing that investigates how short-range dispersal and cell turnover will account for mutational proportions. We show that cancer cells can penetrate neighboring and distinct areas in a matter of days. In next generation sequencing runs, higher proportions of a given cell line generated frequencies with higher precision, while mixtures with lower amounts of each cell line had lower precision manifesting in higher standard deviations. When multiple cell lines were co-cultured, cellular movement altered observed mutation frequency by up to 18.5%. We propose that some of the shared mutations detected at low allele frequencies represent highly motile clones that appear in multiple regions of a tumor owing to dispersion throughout the tumor. In brief, cell movement will lead to a significant technical (sampling) bias when using next generation sequencing to determine clonal composition. A possible solution to this drawback would be to radically decrease detection thresholds and increase coverage in NGS analyses.


Asunto(s)
Heterogeneidad Genética , Variación Genética , Neoplasias/genética , Neoplasias/patología , Línea Celular Tumoral , Movimiento Celular/genética , Biología Computacional/métodos , Progresión de la Enfermedad , Genotipo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Mutación , Invasividad Neoplásica , Metástasis de la Neoplasia
16.
PLoS Comput Biol ; 13(3): e1005411, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28273072

RESUMEN

Mechanical coherence of cell layers is essential for epithelia to function as tissue barriers and to control active tissue dynamics during morphogenesis. RhoA signaling at adherens junctions plays a key role in this process by coupling cadherin-based cell-cell adhesion together with actomyosin contractility. Here we propose and analyze a mathematical model representing core interactions involved in the spatial localization of junctional RhoA signaling. We demonstrate how the interplay between biochemical signaling through positive feedback, combined with diffusion on the cell membrane and mechanical forces generated in the cortex, can determine the spatial distribution of RhoA signaling at cell-cell junctions. This dynamical mechanism relies on the balance between a propagating bistable signal that is opposed by an advective flow generated by an actomyosin stress gradient. Experimental observations on the behavior of the system when contractility is inhibited are in qualitative agreement with the predictions of the model.


Asunto(s)
Actomiosina/fisiología , Uniones Adherentes/fisiología , Células Epiteliales/fisiología , Mecanotransducción Celular/fisiología , Contracción Muscular/fisiología , Proteína de Unión al GTP rhoA/fisiología , Actomiosina/química , Uniones Adherentes/química , Animales , Simulación por Computador , Células Epiteliales/química , Humanos , Modelos Biológicos , Proteínas Motoras Moleculares/química , Proteínas Motoras Moleculares/fisiología , Estrés Mecánico , Proteína de Unión al GTP rhoA/química
17.
Sci Rep ; 6: 17735, 2016 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-26787558

RESUMEN

Cranial neural crest cells (CNCCs) delaminate from embryonic neural folds and migrate to pharyngeal arches, which give rise to most mid-facial structures. CNCC dysfunction plays a prominent role in the etiology of orofacial clefts, a frequent birth malformation. Heterozygous mutations in SPECC1L have been identified in patients with atypical and syndromic clefts. Here, we report that in SPECC1L-knockdown cultured cells, staining of canonical adherens junction (AJ) components, ß-catenin and E-cadherin, was increased, and electron micrographs revealed an apico-basal diffusion of AJs. To understand the role of SPECC1L in craniofacial morphogenesis, we generated a mouse model of Specc1l deficiency. Homozygous mutants were embryonic lethal and showed impaired neural tube closure and CNCC delamination. Staining of AJ proteins was increased in the mutant neural folds. This AJ defect is consistent with impaired CNCC delamination, which requires AJ dissolution. Further, PI3K-AKT signaling was reduced and apoptosis was increased in Specc1l mutants. In vitro, moderate inhibition of PI3K-AKT signaling in wildtype cells was sufficient to cause AJ alterations. Importantly, AJ changes induced by SPECC1L-knockdown were rescued by activating the PI3K-AKT pathway. Together, these data indicate SPECC1L as a novel modulator of PI3K-AKT signaling and AJ biology, required for neural tube closure and CNCC delamination.


Asunto(s)
Uniones Adherentes/metabolismo , Cresta Neural/embriología , Cresta Neural/metabolismo , Fosfoproteínas/deficiencia , Animales , Apoptosis/genética , Biomarcadores , Moléculas de Adhesión Celular/metabolismo , Linaje de la Célula/genética , Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Ratones , Modelos Biológicos , Mutación , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/patología , Fenotipo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
18.
Exp Cell Res ; 319(20): 3094-103, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-23973668

RESUMEN

The mortality of patients with solid tumors is mostly due to metastasis that relies on the interplay between migration and proliferation. The "go or grow" hypothesis postulates that migration and proliferation spatiotemporally excludes each other. We evaluated this hypothesis on 35 cell lines (12 mesothelioma, 13 melanoma and 10 lung cancer) on both the individual cell and population levels. Following three-day-long videomicroscopy, migration, proliferation and cytokinesis-length were quantified. We found a significantly higher migration in mesothelioma cells compared to melanoma and lung cancer while tumor types did not differ in mean proliferation or duration of cytokinesis. Strikingly, we found in melanoma and lung cancer a significant positive correlation between mean proliferation and migration. Furthermore, non-dividing melanoma and lung cancer cells displayed slower migration. In contrast, in mesothelioma there were no such correlations. Interestingly, negative correlation was found between cytokinesis-length and migration in melanoma. FAK activation was higher in melanoma cells with high motility. We demonstrate that the cancer cells studied do not defer proliferation for migration. Of note, tumor cells from various organ systems may differently regulate migration and proliferation. Furthermore, our data is in line with the observation of pathologists that highly proliferative tumors are often highly invasive.


Asunto(s)
Movimiento Celular , Citocinesis , Modelos Biológicos , Neoplasias/patología , Proliferación Celular , Humanos , Células Tumorales Cultivadas
19.
Dev Biol ; 358(2): 331-43, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21871877

RESUMEN

The epicardium is a major contributor of the cells that are required for the formation of coronary vessels. Mice lacking both copies of the gene encoding the Type III Transforming Growth Factor ß Receptor (TGFßR3) fail to form the coronary vasculature, but the molecular mechanism by which TGFßR3 signals coronary vessel formation is unknown. We used intact embryos and epicardial cells from E11.5 mouse embryos to reveal the mechanisms by which TGFßR3 signals and regulates epicardial cell behavior. Analysis of E13.5 embryos reveals a lower rate of epicardial cell proliferation and decreased epicardially derived cell invasion in Tgfbr3(-/-) hearts. Tgfbr3(-/-) epicardial cells in vitro show decreased proliferation and decreased invasion in response to TGFß1 and TGFß2. Unexpectedly, loss of TGFßR3 also decreases responsiveness to two other important regulators of epicardial cell behavior, FGF2 and HMW-HA. Restoring full length TGFßR3 in Tgfbr3(-/-) cells rescued deficits in invasion in vitro in response TGFß1 and TGFß2 as well as FGF2 and HMW-HA. Expression of TGFßR3 missing the 3 C-terminal amino acids that are required to interact with the scaffolding protein GIPC1 did not rescue any of the deficits. Overexpression of GIPC1 alone in Tgfbr3(-/-) cells did not rescue invasion whereas knockdown of GIPC1 in Tgfbr3(+/+) cells decreased invasion in response to TGFß2, FGF2, and HMW-HA. We conclude that TGFßR3 interaction with GIPC1 is critical for regulating invasion and growth factor responsiveness in epicardial cells and that dysregulation of epicardial cell proliferation and invasion contributes to failed coronary vessel development in Tgfbr3(-/-) mice.


Asunto(s)
Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Neuropéptidos/química , Neuropéptidos/metabolismo , Pericardio/citología , Pericardio/metabolismo , Proteoglicanos/química , Proteoglicanos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/química , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Secuencia de Bases , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Proliferación Celular/efectos de los fármacos , Anomalías de los Vasos Coronarios/embriología , Anomalías de los Vasos Coronarios/genética , Anomalías de los Vasos Coronarios/metabolismo , Cartilla de ADN/genética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Cardiovasculares , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Neuropéptidos/antagonistas & inhibidores , Neuropéptidos/genética , Pericardio/embriología , Embarazo , Dominios y Motivos de Interacción de Proteínas , Proteoglicanos/deficiencia , Proteoglicanos/genética , Receptores de Factores de Crecimiento Transformadores beta/deficiencia , Receptores de Factores de Crecimiento Transformadores beta/genética , Transducción de Señal , Imagen de Lapso de Tiempo , Factor de Crecimiento Transformador beta1/farmacología , Factor de Crecimiento Transformador beta2/farmacología
20.
Cytoskeleton (Hoboken) ; 68(6): 325-39, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21634026

RESUMEN

We investigate the effect of myosin II inhibition on cell shape and nuclear motility in cultures of mouse radial glia-like neural progenitor and rat glioma C6 cells. Instead of reducing nucleokinesis, the myosin II inhibitor blebbistatin provokes an elongated bipolar morphology and increased nuclear motility in both cell types. When myosin II is active, time-resolved traction force measurements indicate a pulling force between the leading edge and the nucleus of C6 cells. In the absence of myosin II activity, traction forces during nucleokinesis are diminished below the sensitivity threshold of our assay. By visualizing the centrosome position in C6 cells with GFP-centrin, we show that in the presence or absence of myosin II activity, the nucleus tends to overtake or lag behind the centrosome, respectively. We interpret these findings with the help of a simple viscoelastic model of the cytoskeleton consisting active contractile and passive compressed elements.


Asunto(s)
Núcleo Celular/metabolismo , Forma de la Célula , Miosina Tipo II/antagonistas & inhibidores , Miosina Tipo II/metabolismo , Actinas/metabolismo , Animales , Núcleo Celular/ultraestructura , Polaridad Celular , Células Cultivadas , Centrosoma/metabolismo , Citoesqueleto/metabolismo , Elasticidad , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Ratones , Microtúbulos/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Neuroglía/citología , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Ratas , Estrés Mecánico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA