Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Mater ; 20(6): 892-903, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33495631

RESUMEN

The basement membrane (BM) is a special type of extracellular matrix and presents the major barrier cancer cells have to overcome multiple times to form metastases. Here we show that BM stiffness is a major determinant of metastases formation in several tissues and identify netrin-4 (Net4) as a key regulator of BM stiffness. Mechanistically, our biophysical and functional analyses in combination with mathematical simulations show that Net4 softens the mechanical properties of native BMs by opening laminin node complexes, decreasing cancer cell potential to transmigrate this barrier despite creating bigger pores. Our results therefore reveal that BM stiffness is dominant over pore size, and that the mechanical properties of 'normal' BMs determine metastases formation and patient survival independent of cancer-mediated alterations. Thus, identifying individual Net4 protein levels within native BMs in major metastatic organs may have the potential to define patient survival even before tumour formation. The ratio of Net4 to laminin molecules determines BM stiffness, such that the more Net4, the softer the BM, thereby decreasing cancer cell invasion activity.


Asunto(s)
Membrana Basal/metabolismo , Fenómenos Mecánicos , Metástasis de la Neoplasia , Fenómenos Biomecánicos , Línea Celular Tumoral , Humanos , Netrinas/metabolismo
2.
EMBO J ; 33(21): 2458-72, 2014 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-25168639

RESUMEN

The urokinase-type plasminogen activator receptor (uPAR) is a non-integrin vitronectin (VN) cell adhesion receptor linked to the plasma membrane by a glycolipid anchor. Through structure-function analyses of uPAR, VN and integrins, we document that uPAR-mediated cell adhesion to VN triggers a novel type of integrin signalling that is independent of integrin-matrix engagement. The signalling is fully active on VN mutants deficient in integrin binding site and is also efficiently transduced by integrins deficient in ligand binding. Although integrin ligation is dispensable, signalling is crucially dependent upon an active conformation of the integrin and its association with intracellular adaptors such as talin. This non-canonical integrin signalling is not restricted to uPAR as it poses no structural constraints to the receptor mediating cell attachment. In contrast to canonical integrin signalling, where integrins form direct mechanical links between the ECM and the cytoskeleton, the molecular mechanism enabling the crosstalk between non-integrin adhesion receptors and integrins is dependent upon membrane tension. This suggests that for this type of signalling, the membrane represents a critical component of the molecular clutch.


Asunto(s)
Integrinas/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Transducción de Señal/fisiología , Vitronectina/metabolismo , Adhesión Celular/fisiología , Células HEK293 , Humanos , Integrinas/genética , Mutación , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Vitronectina/genética
3.
EMBO Rep ; 16(10): 1394-408, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26323721

RESUMEN

Cancer-associated fibroblasts (CAFs) interact with tumour cells and promote growth and metastasis. Here, we show that CAF activation is reversible: chronic hypoxia deactivates CAFs, resulting in the loss of contractile force, reduced remodelling of the surrounding extracellular matrix and, ultimately, impaired CAF-mediated cancer cell invasion. Hypoxia inhibits prolyl hydroxylase domain protein 2 (PHD2), leading to hypoxia-inducible factor (HIF)-1α stabilisation, reduced expression of αSMA and periostin, and reduced myosin II activity. Loss of PHD2 in CAFs phenocopies the effects of hypoxia, which can be prevented by simultaneous depletion of HIF-1α. Treatment with the PHD inhibitor DMOG in an orthotopic breast cancer model significantly decreases spontaneous metastases to the lungs and liver, associated with decreased tumour stiffness and fibroblast activation. PHD2 depletion in CAFs co-injected with tumour cells similarly prevents CAF-induced metastasis to lungs and liver. Our data argue that reversion of CAFs towards a less active state is possible and could have important clinical implications.


Asunto(s)
Hipoxia de la Célula , Fibroblastos/fisiología , Prolina Dioxigenasas del Factor Inducible por Hipoxia/deficiencia , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Neoplasias Mamarias Experimentales/patología , Metástasis de la Neoplasia , Células del Estroma/fisiología , Aminoácidos Dicarboxílicos/farmacología , Animales , Mama/citología , Moléculas de Adhesión Celular/genética , Movimiento Celular/efectos de los fármacos , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/antagonistas & inhibidores , Neoplasias Mamarias Experimentales/fisiopatología , Ratones , Miosina Tipo II/metabolismo , Invasividad Neoplásica , Técnicas de Cultivo de Órganos , Células Tumorales Cultivadas
4.
Nucleic Acids Res ; 41(10): 5400-12, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23580553

RESUMEN

Uncontrolled cell proliferation and cytoskeletal remodeling are responsible for tumor development and ultimately metastasis. A number of studies have implicated microRNAs in the regulation of cancer cell invasion and migration. Here, we show that miR-23b regulates focal adhesion, cell spreading, cell-cell junctions and the formation of lamellipodia in breast cancer (BC), implicating a central role for it in cytoskeletal dynamics. Inhibition of miR-23b, using a specific sponge construct, leads to an increase of cell migration and metastatic spread in vivo, indicating it as a metastatic suppressor microRNA. Clinically, low miR-23b expression correlates with the development of metastases in BC patients. Mechanistically, miR-23b is able to directly inhibit a number of genes implicated in cytoskeletal remodeling in BC cells. Through intracellular signal transduction, growth factors activate the transcription factor AP-1, and we show that this in turn reduces miR-23b levels by direct binding to its promoter, releasing the pro-invasive genes from translational inhibition. In aggregate, miR-23b expression invokes a sophisticated interaction network that co-ordinates a wide range of cellular responses required to alter the cytoskeleton during cancer cell motility.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Movimiento Celular , Citoesqueleto/metabolismo , Regulación Neoplásica de la Expresión Génica , MicroARNs/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Miosinas Cardíacas/metabolismo , Adhesión Celular , Línea Celular , Línea Celular Tumoral , Movimiento Celular/genética , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Femenino , Adhesiones Focales/ultraestructura , Humanos , Ratones , Ratones Desnudos , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Cadenas Ligeras de Miosina/metabolismo , Metástasis de la Neoplasia , Fosforilación , Regiones Promotoras Genéticas , Seudópodos/ultraestructura , Factor de Transcripción AP-1/metabolismo , Transcripción Genética , Quinasas p21 Activadas/metabolismo
5.
Carcinogenesis ; 35(2): 396-406, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24072773

RESUMEN

Protein kinase C iota (PKCι), a serine/threonine kinase required for cell polarity, proliferation and migration, is commonly up- or downregulated in cancer. PKCι is a human oncogene but whether this is related to its role in cell polarity and what repertoire of oncogenes acts in concert with PKCι is not known. We developed a panel of candidate oncogene expressing Madin-Darby canine kidney (MDCK) cells and demonstrated that H-Ras, ErbB2 and phosphatidylinositol 3-kinase transformation led to non-polar spheroid morphogenesis (dysplasia), whereas MDCK spheroids expressing c-Raf or v-Src were largely polarized. We show that small interfering RNA (siRNA)-targeting PKCι decreased the size of all spheroids tested and partially reversed the aberrant polarity phenotype in H-Ras and ErbB2 spheroids only. This indicates distinct requirements for PKCι and moreover that different thresholds of PKCι activity are required for these phenotypes. By manipulating PKCι function using mutant constructs, siRNA depletion or chemical inhibition, we have demonstrated that PKCι is required for polarization of parental MDCK epithelial cysts in a 3D matrix and that there is a threshold of PKCι activity above and below which, disorganized epithelial morphogenesis results. Furthermore, treatment with a novel PKCι inhibitor, CRT0066854, was able to restore polarized morphogenesis in the dysplastic H-Ras spheroids. These results show that tightly regulated PKCι is required for normal-polarized morphogenesis in mammalian cells and that H-Ras and ErbB2 cooperate with PKCι for loss of polarization and dysplasia. The identification of a PKCι inhibitor that can restore polarized morphogenesis has implications for the treatment of Ras and ErbB2 driven malignancies.


Asunto(s)
Polaridad Celular , Transformación Celular Neoplásica/patología , Quistes/patología , Células Epiteliales/patología , Isoenzimas/metabolismo , Morfogénesis/fisiología , Proteína Quinasa C/metabolismo , Esferoides Celulares/patología , Animales , Transformación Celular Neoplásica/metabolismo , Células Cultivadas , Quistes/metabolismo , Perros , Células Epiteliales/metabolismo , Genes ras/fisiología , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/genética , Riñón/metabolismo , Riñón/patología , Fosfatidilinositol 3-Quinasa/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/genética , ARN Interferente Pequeño/genética , Receptor ErbB-2/metabolismo , Esferoides Celulares/metabolismo
6.
J Cell Biol ; 177(5): 927-39, 2007 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-17548516

RESUMEN

Expression of the membrane receptor uPAR induces profound changes in cell morphology and migration, and its expression correlates with the malignant phenotype of cancers. To identify the molecular interactions essential for uPAR function in these processes, we carried out a complete functional alanine scan of uPAR in HEK293 cells. Of the 255 mutant receptors characterized, 34 failed to induce changes in cell morphology. Remarkably, the molecular defect of all of these mutants was a specific reduction in integrin-independent cell binding to vitronectin. A membrane-tethered plasminogen activator inhibitor-1, which has the same binding site in vitronectin as uPAR, replicated uPAR-induced changes. A direct uPAR-vitronectin interaction is thus both required and sufficient to initiate downstream changes in cell morphology, migration, and signal transduction. Collectively these data demonstrate a novel mechanism by which a cell adhesion molecule lacking inherent signaling capability evokes complex cellular responses by modulating the contact between the cell and the matrix without the requirement for direct lateral protein-protein interactions.


Asunto(s)
Movimiento Celular/fisiología , Receptores de Superficie Celular/fisiología , Vitronectina/fisiología , Secuencias de Aminoácidos , Animales , Sitios de Unión , Células CHO , Adhesión Celular/fisiología , Línea Celular , Cricetinae , Cricetulus , Prueba de Complementación Genética , Humanos , Mutagénesis Sitio-Dirigida , Mapeo de Interacción de Proteínas , Receptores de Superficie Celular/química , Receptores de Superficie Celular/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Transducción de Señal , Vitronectina/química , Vitronectina/metabolismo
7.
Sci Transl Med ; 14(666): eabm6391, 2022 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-36223446

RESUMEN

The bone marrow microenvironment provides indispensable factors to sustain blood production throughout life. It is also a hotspot for the progression of hematologic disorders and the most frequent site of solid tumor metastasis. Preclinical research relies on xenograft mouse models, but these models preclude the human-specific functional interactions of stem cells with their bone marrow microenvironment. Instead, human mesenchymal cells can be exploited for the in vivo engineering of humanized niches, which confer robust engraftment of human healthy and malignant blood samples. However, mesenchymal cells are associated with major reproducibility issues in tissue formation. Here, we report the fast and standardized generation of human mini-bones by a custom-designed human mesenchymal cell line. These resulting humanized ossicles (hOss) consist of fully mature bone and bone marrow structures hosting a human mesenchymal niche with retained stem cell properties. As compared to mouse bones, we demonstrate superior engraftment of human cord blood hematopoietic cells and primary acute myeloid leukemia samples and also validate hOss as a metastatic site for breast cancer cells. We further report the engraftment of neuroblastoma patient-derived xenograft cells in a humanized model, recapitulating clinically described osteolytic lesions. Collectively, our human mini-bones constitute a powerful preclinical platform to model bone-developing tumors using patient-derived materials.


Asunto(s)
Leucemia Mieloide Aguda , Nicho de Células Madre , Animales , Huesos , Modelos Animales de Enfermedad , Hematopoyesis , Humanos , Ratones , Reproducibilidad de los Resultados , Microambiente Tumoral
8.
Adv Healthc Mater ; 11(1): e2100684, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34734500

RESUMEN

Metastatic cancer spread is responsible for most cancer-related deaths. To colonize a new organ, invading cells adapt to, and remodel, the local extracellular matrix (ECM), a network of proteins and proteoglycans underpinning all tissues, and a critical regulator of homeostasis and disease. However, there is a major lack in tools to study cancer cell behavior within native 3D ECM. Here, an in-house designed bioreactor, where mouse organ ECM scaffolds are perfused and populated with cells that are challenged to colonize it, is presented. Using a specialized bioreactor chamber, it is possible to monitor cell behavior microscopically (e.g., proliferation, migration) within the organ scaffold. Cancer cells in this system recapitulate cell signaling observed in vivo and remodel complex native ECM. Moreover, the bioreactors are compatible with co-culturing cell types of different genetic origin comprising the normal and tumor microenvironment. This degree of experimental flexibility in an organ-specific and 3D context, opens new possibilities to study cell-cell and cell-ECM interplay and to model diseases in a controllable organ-specific system ex vivo.


Asunto(s)
Matriz Extracelular , Andamios del Tejido , Animales , Reactores Biológicos , Ratones , Perfusión , Proteoglicanos , Ingeniería de Tejidos
9.
Cancer Cell ; 39(4): 451-453, 2021 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-33711271

RESUMEN

Clinical implementation of anti-stromal therapies in pancreatic cancer has been delayed by unanticipated tumor-restraining properties of the desmoplastic stroma. In confronting these challenges, Chen et al. demonstrate in this issue of Cancer Cell that fibroblast-specific deletion of collagen I, in the background of oncogenic Kras-induced spontaneous murine pancreatic ductal adenocarcinoma, enhances immune suppression and accelerates progression of disease.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Animales , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Colágeno , Fibroblastos , Ratones , Conductos Pancreáticos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética
10.
J Vis Exp ; (171)2021 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-34125099

RESUMEN

We present here a decellularization protocol for mouse heart and lungs. It produces structural ECM scaffolds that can be used to analyze ECM topology and composition. It is based on a microsurgical procedure designed to catheterize the trachea and aorta of a euthanized mouse to perfuse the heart and lungs with decellularizing agents. The decellularized cardiopulmonary complex can subsequently be immunostained to reveal the location of structural ECM proteins. The whole procedure can be completed in 4 days. The ECM scaffolds resulting from this protocol are free of dimensional distortions. The absence of cells enables structural examination of ECM structures down to submicron resolution in 3D. This protocol can be applied to healthy and diseased tissue from mice as young as 4-weeks old, including mouse models of fibrosis and cancer, opening the way to determine ECM remodeling associated with cardiopulmonary disease.


Asunto(s)
Corazón , Pulmón , Animales , Matriz Extracelular , Ratones , Ingeniería de Tejidos , Andamios del Tejido
11.
Nat Cell Biol ; 23(7): 758-770, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34226698

RESUMEN

The YAP/TAZ transcriptional programme is not only a well-established driver of cancer progression and metastasis but also an important stimulator of tissue regeneration. Here we identified Cerebral cavernous malformations 3 (CCM3) as a regulator of mechanical cue-driven YAP/TAZ signalling, controlling both tumour progression and stem cell differentiation. We demonstrate that CCM3 localizes to focal adhesion sites in cancer-associated fibroblasts, where it regulates mechanotransduction and YAP/TAZ activation. Mechanistically, CCM3 and focal adhesion kinase (FAK) mutually compete for binding to paxillin to fine-tune FAK/Src/paxillin-driven mechanotransduction and YAP/TAZ activation. In mouse models of breast cancer, specific loss of CCM3 in cancer-associated fibroblasts leads to exacerbated tissue remodelling and force transmission to the matrix, resulting in reciprocal YAP/TAZ activation in the neighbouring tumour cells and dissemination of metastasis to distant organs. Similarly, CCM3 regulates the differentiation of mesenchymal stromal/stem cells. In conclusion, CCM3 is a gatekeeper in focal adhesions that controls mechanotransduction and YAP/TAZ signalling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Neoplasias de la Mama/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Adhesiones Focales/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Mecanotransducción Celular , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas Reguladoras de la Apoptosis/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Fibroblastos Asociados al Cáncer/patología , Comunicación Celular , Diferenciación Celular , Línea Celular Tumoral , Femenino , Quinasa 1 de Adhesión Focal/metabolismo , Adhesiones Focales/genética , Adhesiones Focales/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Metástasis de la Neoplasia , Paxillin/metabolismo , Fosforilación , Unión Proteica , Proteínas Proto-Oncogénicas/genética , Estrés Mecánico , Factores de Transcripción/genética , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Proteínas Señalizadoras YAP , Familia-src Quinasas/metabolismo
13.
Front Cell Dev Biol ; 8: 146, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32258031

RESUMEN

The STRIPAK complex has been linked to a variety of biological processes taking place during embryogenesis and development, but its role in cancer has only just started to be defined. Here, we expand on previous work indicating a role for the scaffolding protein STRIP1 in cancer cell migration and metastasis. We show that cell cycle arrest and decreased proliferation are seen upon loss of STRIP1 in MDA-MB-231 cells due to the induction of cyclin dependent kinase inhibitors, including p21 and p27. We demonstrate that p21 and p27 induction is observed in a subpopulation of cells having low DNA damage response and that the p21high/γH2AXlow ratio within single cells can be rescued by depleting MST3&4 kinases. While the loss of STRIP1 decreases cell proliferation and tumor growth, cells treated with low dosage of chemotherapeutics in vitro paradoxically escape therapy-induced senescence and begin to proliferate after recovery. This corroborates with already known research on the dual role of p21 and indicates that STRIP1 also plays a contradictory role in breast cancer, suppressing tumor growth, but once treated with chemotherapeutics, allowing for possible recurrence and decreased patient survival.

14.
Cancer Rep (Hoboken) ; 3(1): e1209, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32671954

RESUMEN

BACKGROUND: The use of in vitro cell cultures is a powerful tool for obtaining key insights into the behaviour and response of cells to interventions in normal and disease situations. Unlike in vivo settings, in vitro experiments allow a fine-tuned control of a range of microenvironmental elements independently within an isolated setting. The recent expansion in the use of three-dimensional (3D) in vitro assays has created a number of representative tools to study cell behaviour in a more physiologically 3D relevant microenvironment. Complex 3D in vitro models that can recapitulate human tissue biology are essential for understanding the pathophysiology of disease. AIM: The development of the 3D coculture collagen contraction and invasion assay, the "organotypic assay," has been widely adopted as a powerful approach to bridge the gap between standard two-dimensional tissue culture and in vivo mouse models. In the cancer setting, these assays can then be used to dissect how stromal cells, such as cancer-associated fibroblasts (CAFs), drive extracellular matrix (ECM) remodelling to alter cancer cell behaviour and response to intervention. However, to date, many of the published organotypic protocols are low-throughput, time-consuming (up to several weeks), and work-intensive with often limited scalability. Our aim was to develop a fast, high-throughput, scalable 3D organotypic assay for use in oncology screening and drug development. METHODS AND RESULTS: Here, we describe a modified 96-well organotypic assay, the "Mini-Organo," which can be easily completed within 5 days. We demonstrate its application in a wide range of mouse and human cancer biology approaches including evaluation of stromal cell 3D ECM remodelling, 3D cancer cell invasion, and the assessment of efficacy of potential anticancer therapeutic targets. Furthermore, the organotypic assay described is highly amenable to customisation using different cell types under diverse experimental conditions. CONCLUSIONS: The Mini-Organo high-throughput 3D organotypic assay allows the rapid screening of potential cancer therapeutics in human and mouse models in a time-efficient manner.


Asunto(s)
Técnicas de Cocultivo/métodos , Desarrollo de Medicamentos/métodos , Ensayos de Selección de Medicamentos Antitumorales/métodos , Animales , Fibroblastos Asociados al Cáncer/fisiología , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones , Invasividad Neoplásica , Ratas
15.
Nat Protoc ; 14(12): 3395-3425, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31705125

RESUMEN

The extracellular matrix (ECM) is a major regulator of homeostasis and disease, yet the 3D structure of the ECM remains poorly understood because of limitations in ECM visualization. We recently developed an ECM-specialized method termed in situ decellularization of tissues (ISDoT) to isolate native 3D ECM scaffolds from whole organs in which ECM structure and composition are preserved. Here, we present detailed surgical instructions to facilitate decellularization of 33 different mouse tissues and details of validated antibodies that enable the visualization of 35 mouse ECM proteins. Through mapping of these ECM proteins, the structure of the ECM can be determined and tissue structures visualized in detail. In this study, perfusion decellularization is presented for bones, skeletal muscle, tongue, salivary glands, stomach, duodenum, jejunum/ileum, large intestines, mesentery, liver, gallbladder, pancreas, trachea, bronchi, lungs, kidneys, urinary bladder, ovaries, uterine horn, cervix, adrenal gland, heart, arteries, veins, capillaries, lymph nodes, spleen, peripheral nerves, eye, outer ear, mammary glands, skin, and subcutaneous tissue. Decellularization, immunostaining, and imaging take 4-5 d.


Asunto(s)
Matriz Extracelular/metabolismo , Imagenología Tridimensional/métodos , Coloración y Etiquetado/métodos , Animales , Anticuerpos/metabolismo , Matriz Extracelular/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Especificidad de Órganos , Perfusión/métodos , Ingeniería de Tejidos/métodos , Andamios del Tejido/química
16.
Eur J Cell Biol ; 87(8-9): 617-29, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18353489

RESUMEN

The extracellular matrix (ECM) is a complex structural entity surrounding and supporting cells present in all tissue and organs. Cell-matrix interactions play fundamental roles during embryonic development, morphogenesis, tissue homoeostasis, wound healing, and tumourigenesis. Cell-matrix communication is kept in balance by physical contact and by transmembrane integrin receptors providing the dynamic link between the extracellular and intracellular environments through bi-directional signalling. The urokinase-type plasminogen activator receptor (uPAR) is a plasma membrane receptor overexpressed during inflammation and in almost all human cancers. One of its functions is to endorse ECM remodelling through the activation of plasminogen and downstream proteases, including matrix-metalloproteases (MMPs). Beside its role in ECM degradation, uPAR modulates cell-matrix contact through a direct engagement with the ECM component, vitronectin (Vn), and by regulating the activity state of integrins thus promoting or inhibiting integrin signalling and integrin-mediated cell adhesion to other ECM components, like fibronectin and collagen. In this review we have centred our attention on the non-proteolytic function of uPAR as a mediator of cell adhesion and downstream signalling.


Asunto(s)
Adhesión Celular , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Vitronectina/metabolismo , Animales , Matriz Extracelular/metabolismo , Adhesiones Focales , VIH-1/metabolismo , Humanos , Integrinas/metabolismo , Modelos Biológicos , Receptores del Activador de Plasminógeno Tipo Uroquinasa
17.
Bio Protoc ; 8(6): e2776, 2018 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-34179290

RESUMEN

Mesenchymal stem cells (MSCs) have shown profound therapeutic potential in tissue repair and regeneration. However, recent studies indicate that MSCs are largely entrapped in lungs after intravenous delivery and die shortly. The underlying mechanisms have been poorly understood. We have provided evidence to show that excess expression and activation of integrins in culture-expanded MSCs is a critical cause of MSCs adhesion to endothelial cells of the lung microarteries resulting in the entrapment of the cells ( Wang et al., 2015 ). Therefore, it may be meaningful to test the adhesive ability of MSCs to endothelial cells in vitro before intravenous administration to avoid their lung vascular obstructions. Here we report a simple method to measure MSCs attachment to endothelial cells.

18.
Nat Commun ; 9(1): 5150, 2018 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-30514914

RESUMEN

Cancer-associated fibroblasts (CAFs) are a major constituent of the tumor microenvironment, although their origin and roles in shaping disease initiation, progression and treatment response remain unclear due to significant heterogeneity. Here, following a negative selection strategy combined with single-cell RNA sequencing of 768 transcriptomes of mesenchymal cells from a genetically engineered mouse model of breast cancer, we define three distinct subpopulations of CAFs. Validation at the transcriptional and protein level in several experimental models of cancer and human tumors reveal spatial separation of the CAF subclasses attributable to different origins, including the peri-vascular niche, the mammary fat pad and the transformed epithelium. Gene profiles for each CAF subtype correlate to distinctive functional programs and hold independent prognostic capability in clinical cohorts by association to metastatic disease. In conclusion, the improved resolution of the widely defined CAF population opens the possibility for biomarker-driven development of drugs for precision targeting of CAFs.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Fibroblastos Asociados al Cáncer , Análisis de Secuencia de ARN , Transcriptoma , Tejido Adiposo/metabolismo , Animales , Biomarcadores de Tumor/genética , Mama/metabolismo , Mama/patología , Neoplasias de la Mama/patología , Fibroblastos Asociados al Cáncer/clasificación , Ciclo Celular/genética , Línea Celular Tumoral , Análisis por Conglomerados , Progresión de la Enfermedad , Epitelio/metabolismo , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Uniones Intercelulares/genética , Modelos Logísticos , Ratones , Ratones Transgénicos , Pronóstico , Factores de Transcripción/genética , Transcriptoma/genética
19.
Mol Biol Cell ; 29(20): 2378-2385, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30091653

RESUMEN

Increased tissue stiffness is a classic characteristic of solid tumors. One of the major contributing factors is increased density of collagen fibers in the extracellular matrix (ECM). Here, we investigate how cancer cells biomechanically interact with and respond to the stiffness of the ECM. Probing the adaptability of cancer cells to altered ECM stiffness using optical tweezers-based microrheology and deformability cytometry, we find that only malignant cancer cells have the ability to adjust to collagen matrices of different densities. Employing microrheology on the biologically relevant spheroid invasion assay, we can furthermore demonstrate that, even within a cluster of cells of similar origin, there are differences in the intracellular biomechanical properties dependent on the cells' invasive behavior. We reveal a consistent increase of viscosity in cancer cells leading the invasion into the collagen matrices in comparison with cancer cells following in the stalk or remaining in the center of the spheroid. We hypothesize that this differential viscoelasticity might facilitate spheroid tip invasion through a dense matrix. These findings highlight the importance of the biomechanical interplay between cells and their microenvironment for tumor progression.


Asunto(s)
Matriz Extracelular/metabolismo , Neoplasias/patología , Animales , Fenómenos Biomecánicos , Línea Celular Tumoral , Elasticidad , Humanos , Ratones , Invasividad Neoplásica , Ratas , Reología , Viscosidad
20.
Cell Stem Cell ; 22(1): 35-49.e7, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29249464

RESUMEN

Tissue regeneration requires dynamic cellular adaptation to the wound environment. It is currently unclear how this is orchestrated at the cellular level and how cell fate is affected by severe tissue damage. Here we dissect cell fate transitions during colonic regeneration in a mouse dextran sulfate sodium (DSS) colitis model, and we demonstrate that the epithelium is transiently reprogrammed into a primitive state. This is characterized by de novo expression of fetal markers as well as suppression of markers for adult stem and differentiated cells. The fate change is orchestrated by remodeling the extracellular matrix (ECM), increased FAK/Src signaling, and ultimately YAP/TAZ activation. In a defined cell culture system recapitulating the extracellular matrix remodeling observed in vivo, we show that a collagen 3D matrix supplemented with Wnt ligands is sufficient to sustain endogenous YAP/TAZ and induce conversion of cell fate. This provides a simple model for tissue regeneration, implicating cellular reprogramming as an essential element.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Reprogramación Celular , Matriz Extracelular/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Fosfoproteínas/metabolismo , Regeneración , Animales , Biomarcadores/metabolismo , Proteínas de Ciclo Celular , Feto/metabolismo , Humanos , Mecanotransducción Celular , Ratones Endogámicos C57BL , Transducción de Señal , Transcripción Genética , Activación Transcripcional/genética , Proteínas Señalizadoras YAP
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA