Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 154
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 187(12): 3072-3089.e20, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38781967

RESUMEN

Tissue folds are structural motifs critical to organ function. In the intestine, bending of a flat epithelium into a periodic pattern of folds gives rise to villi, finger-like protrusions that enable nutrient absorption. However, the molecular and mechanical processes driving villus morphogenesis remain unclear. Here, we identify an active mechanical mechanism that simultaneously patterns and folds the intestinal epithelium to initiate villus formation. At the cellular level, we find that PDGFRA+ subepithelial mesenchymal cells generate myosin II-dependent forces sufficient to produce patterned curvature in neighboring tissue interfaces. This symmetry-breaking process requires altered cell and extracellular matrix interactions that are enabled by matrix metalloproteinase-mediated tissue fluidization. Computational models, together with in vitro and in vivo experiments, revealed that these cellular features manifest at the tissue level as differences in interfacial tensions that promote mesenchymal aggregation and interface bending through a process analogous to the active dewetting of a thin liquid film.


Asunto(s)
Matriz Extracelular , Mucosa Intestinal , Animales , Ratones , Mucosa Intestinal/metabolismo , Mucosa Intestinal/citología , Matriz Extracelular/metabolismo , Miosina Tipo II/metabolismo , Mesodermo/metabolismo , Mesodermo/citología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Morfogénesis , Metaloproteinasas de la Matriz/metabolismo
2.
Annu Rev Cell Dev Biol ; 32: 527-554, 2016 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-27298087

RESUMEN

Mechanical force modulates development, influences tissue homeostasis, and contributes to disease. Forces sculpt tissue-level behaviors and direct cell fate by engaging mechanoreceptors and by altering organization of the cytoskeleton and actomyosin contractility to stimulate mechanotransduction mechanisms that alter transcription. Nevertheless, how force specifically leverages mechanotransduction pathways to control transcriptional regulation of cell fate remains unclear. Here we review recent findings specifically in the context of epithelial-to-mesenchymal transitions that have revealed conserved mechanisms whereby extracellular force, mediated through cell-extracellular matrix and cell-cell junctional complexes, induces transcriptional reprogramming to alter cell and tissue fate. We focus on the interplay between tissue mechanics and the epithelial-to-mesenchymal transitions that occur during embryonic development and cancer malignancy. We describe the adhesion-linked cellular machinery that mediates mechano-transduction and elaborate on how these force-linked networks stimulate key transcriptional programs that induce an epithelial-to-mesenchymal phenotypic transition, thereby providing an overview of how mechanical signals can be translated into a change in cell fate.


Asunto(s)
Desarrollo Embrionario , Transición Epitelial-Mesenquimal , Mecanotransducción Celular , Neoplasias/patología , Animales , Retroalimentación Fisiológica , Humanos , Transducción de Señal
3.
Genes Dev ; 35(13-14): 963-975, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34168038

RESUMEN

Autophagy inhibitors are currently being evaluated in clinical trials for the treatment of diverse cancers, largely due to their ability to impede tumor cell survival and metabolic adaptation. More recently, there is growing interest in whether and how modulating autophagy in the host stroma influences tumorigenesis. Fibroblasts play prominent roles in cancer initiation and progression, including depositing type 1 collagen and other extracellular matrix (ECM) components, thereby stiffening the surrounding tissue to enhance tumor cell proliferation and survival, as well as secreting cytokines that modulate angiogenesis and the immune microenvironment. This constellation of phenotypes, pathologically termed desmoplasia, heralds poor prognosis and reduces patient survival. Using mouse mammary cancer models and syngeneic transplantation assays, we demonstrate that genetic ablation of stromal fibroblast autophagy significantly impedes fundamental elements of the stromal desmoplastic response, including collagen and proinflammatory cytokine secretion, extracellular matrix stiffening, and neoangiogenesis. As a result, autophagy in stromal fibroblasts is required for mammary tumor growth in vivo, even when the cancer cells themselves remain autophagy-competent . We propose the efficacy of autophagy inhibition is shaped by this ability of host stromal fibroblast autophagy to support tumor desmoplasia.


Asunto(s)
Células del Estroma , Microambiente Tumoral , Animales , Autofagia/genética , Línea Celular Tumoral , Transformación Celular Neoplásica/patología , Fibroblastos/metabolismo , Humanos , Ratones , Microambiente Tumoral/genética
4.
Proc Natl Acad Sci U S A ; 121(20): e2322688121, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38709925

RESUMEN

Brain metastatic breast cancer is particularly lethal largely due to therapeutic resistance. Almost half of the patients with metastatic HER2-positive breast cancer develop brain metastases, representing a major clinical challenge. We previously described that cancer-associated fibroblasts are an important source of resistance in primary tumors. Here, we report that breast cancer brain metastasis stromal cell interactions in 3D cocultures induce therapeutic resistance to HER2-targeting agents, particularly to the small molecule inhibitor of HER2/EGFR neratinib. We investigated the underlying mechanisms using a synthetic Notch reporter system enabling the sorting of cancer cells that directly interact with stromal cells. We identified mucins and bulky glycoprotein synthesis as top-up-regulated genes and pathways by comparing the gene expression and chromatin profiles of stroma-contact and no-contact cancer cells before and after neratinib treatment. Glycoprotein gene signatures were also enriched in human brain metastases compared to primary tumors. We confirmed increased glycocalyx surrounding cocultures by immunofluorescence and showed that mucinase treatment increased sensitivity to neratinib by enabling a more efficient inhibition of EGFR/HER2 signaling in cancer cells. Overexpression of truncated MUC1 lacking the intracellular domain as a model of increased glycocalyx-induced resistance to neratinib both in cell culture and in experimental brain metastases in immunodeficient mice. Our results highlight the importance of glycoproteins as a resistance mechanism to HER2-targeting therapies in breast cancer brain metastases.


Asunto(s)
Neoplasias Encefálicas , Neoplasias de la Mama , Resistencia a Antineoplásicos , Glicocálix , Quinolinas , Receptor ErbB-2 , Células del Estroma , Humanos , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Femenino , Neoplasias Encefálicas/secundario , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/genética , Receptor ErbB-2/metabolismo , Receptor ErbB-2/genética , Glicocálix/metabolismo , Animales , Línea Celular Tumoral , Células del Estroma/metabolismo , Células del Estroma/patología , Quinolinas/farmacología , Ratones , Comunicación Celular , Técnicas de Cocultivo , Mucina-1/metabolismo , Mucina-1/genética , Transducción de Señal , Receptores ErbB/metabolismo , Receptores ErbB/antagonistas & inhibidores
5.
EMBO J ; 41(17): e109205, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35880301

RESUMEN

Patient-derived organoids and cellular spheroids recapitulate tissue physiology with remarkable fidelity. We investigated how engagement with a reconstituted basement membrane in three dimensions (3D) supports the polarized, stress resilient tissue phenotype of mammary epithelial spheroids. Cells interacting with reconstituted basement membrane in 3D had reduced levels of total and actin-associated filamin and decreased cortical actin tension that increased plasma membrane protrusions to promote negative plasma membrane curvature and plasma membrane protein associations linked to protein secretion. By contrast, cells engaging a reconstituted basement membrane in 2D had high cortical actin tension that forced filamin unfolding and endoplasmic reticulum (ER) associations. Enhanced filamin-ER interactions increased levels of PKR-like ER kinase effectors and ER-plasma membrane contact sites that compromised calcium homeostasis and diminished cell viability. Consequently, cells with decreased cortical actin tension had reduced ER stress and survived better. Consistently, cortical actin tension in cellular spheroids regulated polarized basement membrane membrane deposition and sensitivity to exogenous stress. The findings implicate cortical actin tension-mediated filamin unfolding in ER function and underscore the importance of tissue mechanics in organoid homeostasis.


Asunto(s)
Actinas , Retículo Endoplásmico , Actinas/metabolismo , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico , Células Epiteliales/metabolismo , Filaminas/metabolismo , Fenotipo
6.
Nat Rev Mol Cell Biol ; 15(12): 771-85, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25370693

RESUMEN

The biochemical and biophysical properties of the extracellular matrix (ECM) dictate tissue-specific cell behaviour. The molecules that are associated with the ECM of each tissue, including collagens, proteoglycans, laminins and fibronectin, and the manner in which they are assembled determine the structure and the organization of the resultant ECM. The product is a specific ECM signature that is comprised of unique compositional and topographical features that both reflect and facilitate the functional requirements of the tissue.


Asunto(s)
Matriz Extracelular/química , Matriz Extracelular/metabolismo , Animales , Membrana Basal/química , Membrana Basal/metabolismo , Proteínas de la Matriz Extracelular/química , Proteínas de la Matriz Extracelular/metabolismo , Humanos , Neuronas/citología , Proteoglicanos/metabolismo
7.
Genes Dev ; 32(3-4): 244-257, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29483153

RESUMEN

The discoidin domain receptor 1 (DDR1) is overexpressed in breast carcinoma cells. Low DDR1 expression is associated with worse relapse-free survival, reflecting its controversial role in cancer progression. We detected DDR1 on luminal cells but not on myoepithelial cells of DDR1+/+ mice. We found that DDR1 loss compromises cell adhesion, consistent with data that older DDR1-/- mammary glands had more basal/myoepithelial cells. Basal cells isolated from older mice exerted higher traction forces than the luminal cells, in agreement with increased mammary branches observed in older DDR1-/- mice and higher branching by their isolated organoids. When we crossed DDR1-/- mice with MMTV-PyMT mice, the PyMT/DDR1-/- mammary tumors grew faster and had increased epithelial tension and matricellular fibrosis with a more basal phenotype and increased lung metastases. DDR1 deletion induced basal differentiation of CD90+CD24+ cancer cells, and the increase in basal cells correlated with tumor cell mitoses. K14+ basal cells, including K8+K14+ cells, were increased adjacent to necrotic fields. These data suggest that the absence of DDR1 provides a growth and adhesion advantage that favors the expansion of basal cells, potentiates fibrosis, and enhances necrosis/hypoxia and basal differentiation of transformed cells to increase their aggression and metastatic potential.


Asunto(s)
Receptor con Dominio Discoidina 1/genética , Neoplasias Mamarias Experimentales/patología , Animales , Neoplasias de la Mama/metabolismo , Hipoxia de la Célula , Receptor con Dominio Discoidina 1/metabolismo , Supervivencia sin Enfermedad , Células Epiteliales/metabolismo , Femenino , Fibrosis , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/genética , Ratones
8.
Nature ; 573(7774): 421-425, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31511693

RESUMEN

Early human embryonic development involves extensive lineage diversification, cell-fate specification and tissue patterning1. Despite its basic and clinical importance, early human embryonic development remains relatively unexplained owing to interspecies divergence2,3 and limited accessibility to human embryo samples. Here we report that human pluripotent stem cells (hPSCs) in a microfluidic device recapitulate, in a highly controllable and scalable fashion, landmarks of the development of the epiblast and amniotic ectoderm parts of the conceptus, including lumenogenesis of the epiblast and the resultant pro-amniotic cavity, formation of a bipolar embryonic sac, and specification of primordial germ cells and primitive streak cells. We further show that amniotic ectoderm-like cells function as a signalling centre to trigger the onset of gastrulation-like events in hPSCs. Given its controllability and scalability, the microfluidic model provides a powerful experimental system to advance knowledge of human embryology and reproduction. This model could assist in the rational design of differentiation protocols of hPSCs for disease modelling and cell therapy, and in high-throughput drug and toxicity screens to prevent pregnancy failure and birth defects.


Asunto(s)
Amnios/embriología , Estratos Germinativos/embriología , Modelos Biológicos , Células Madre Pluripotentes/citología , Amnios/citología , Diferenciación Celular , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Femenino , Estratos Germinativos/citología , Humanos , Embarazo , Línea Primitiva/citología
9.
Cell ; 139(5): 891-906, 2009 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-19931152

RESUMEN

Tumors are characterized by extracellular matrix (ECM) remodeling and stiffening. The importance of ECM remodeling to cancer is appreciated; the relevance of stiffening is less clear. We found that breast tumorigenesis is accompanied by collagen crosslinking, ECM stiffening, and increased focal adhesions. Induction of collagen crosslinking stiffened the ECM, promoted focal adhesions, enhanced PI3 kinase (PI3K) activity, and induced the invasion of an oncogene-initiated epithelium. Inhibition of integrin signaling repressed the invasion of a premalignant epithelium into a stiffened, crosslinked ECM and forced integrin clustering promoted focal adhesions, enhanced PI3K signaling, and induced the invasion of a premalignant epithelium. Consistently, reduction of lysyl oxidase-mediated collagen crosslinking prevented MMTV-Neu-induced fibrosis, decreased focal adhesions and PI3K activity, impeded malignancy, and lowered tumor incidence. These data show how collagen crosslinking can modulate tissue fibrosis and stiffness to force focal adhesions, growth factor signaling and breast malignancy.


Asunto(s)
Neoplasias de la Mama/patología , Matriz Extracelular/metabolismo , Integrinas/metabolismo , Envejecimiento , Animales , Colágeno/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Femenino , Fibrosis/patología , Genes ras , Humanos , Glándulas Mamarias Humanas/patología , Ratones , Ratones Endogámicos BALB C , Proteína-Lisina 6-Oxidasa/metabolismo , Transducción de Señal
10.
Nat Rev Mol Cell Biol ; 12(5): 308-19, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21508987

RESUMEN

All cells exist within the context of a three-dimensional microenvironment in which they are exposed to mechanical and physical cues. These cues can be disrupted through perturbations to mechanotransduction, from the nanoscale-level to the tissue-level, which compromises tensional homeostasis to promote pathologies such as cardiovascular disease and cancer. The mechanisms of such perturbations suggest that a complex interplay exists between the extracellular microenvironment and cellular function. Furthermore, sustained disruptions in tensional homeostasis can be caused by alterations in the extracellular matrix, allowing it to serve as a mechanically based memory-storage device that can perpetuate a disease or restore normal tissue behaviour.


Asunto(s)
Matriz Extracelular/fisiología , Homeostasis/fisiología , Uniones Intercelulares/fisiología , Mecanotransducción Celular/fisiología , Animales , Adhesión Celular/fisiología , Humanos , Modelos Biológicos , Estrés Mecánico
11.
Nat Mater ; 20(4): 548-559, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33257795

RESUMEN

Stromal stiffening accompanies malignancy, compromises treatment and promotes tumour aggression. Clarifying the molecular nature and the factors that regulate stromal stiffening in tumours should identify biomarkers to stratify patients for therapy and interventions to improve outcome. We profiled lysyl hydroxylase-mediated and lysyl oxidase-mediated collagen crosslinks and quantified the greatest abundance of total and complex collagen crosslinks in aggressive human breast cancer subtypes with the stiffest stroma. These tissues harbour the highest number of tumour-associated macrophages, whose therapeutic ablation in experimental models reduced metastasis, and decreased collagen crosslinks and stromal stiffening. Epithelial-targeted expression of the crosslinking enzyme, lysyl oxidase, had no impact on collagen crosslinking in PyMT mammary tumours, whereas stromal cell targeting did. Stromal cells in microdissected human tumours expressed the highest level of collagen crosslinking enzymes. Immunohistochemical analysis of biopsies from a cohort of patients with breast cancer revealed that stromal expression of lysyl hydroxylase 2, an enzyme that induces hydroxylysine aldehyde-derived collagen crosslinks and stromal stiffening, correlated significantly with disease specific mortality. The findings link tissue inflammation, stromal cell-mediated collagen crosslinking and stiffening to tumour aggression and identify lysyl hydroxylase 2 as a stromal biomarker.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Colágeno/metabolismo , Células del Estroma/metabolismo , Macrófagos Asociados a Tumores/metabolismo , Adulto , Biopsia , Neoplasias de la Mama/inmunología , Línea Celular Tumoral , Femenino , Humanos , Persona de Mediana Edad , Proteína-Lisina 6-Oxidasa/metabolismo , Células del Estroma/patología
12.
Nature ; 578(7796): 517-518, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32094916
14.
Nature ; 511(7509): 319-25, 2014 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-25030168

RESUMEN

Malignancy is associated with altered expression of glycans and glycoproteins that contribute to the cellular glycocalyx. We constructed a glycoprotein expression signature, which revealed that metastatic tumours upregulate expression of bulky glycoproteins. A computational model predicted that these glycoproteins would influence transmembrane receptor spatial organization and function. We tested this prediction by investigating whether bulky glycoproteins in the glycocalyx promote a tumour phenotype in human cells by increasing integrin adhesion and signalling. Our data revealed that a bulky glycocalyx facilitates integrin clustering by funnelling active integrins into adhesions and altering integrin state by applying tension to matrix-bound integrins, independent of actomyosin contractility. Expression of large tumour-associated glycoproteins in non-transformed mammary cells promoted focal adhesion assembly and facilitated integrin-dependent growth factor signalling to support cell growth and survival. Clinical studies revealed that large glycoproteins are abundantly expressed on circulating tumour cells from patients with advanced disease. Thus, a bulky glycocalyx is a feature of tumour cells that could foster metastasis by mechanically enhancing cell-surface receptor function.


Asunto(s)
Glicocálix/metabolismo , Glicoproteínas/metabolismo , Integrinas/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Animales , Mama/citología , Mama/metabolismo , Mama/patología , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Fibroblastos , Glicocálix/química , Humanos , Proteínas Inmovilizadas/química , Proteínas Inmovilizadas/metabolismo , Integrinas/química , Ratones , Terapia Molecular Dirigida , Mucina-1/metabolismo , Metástasis de la Neoplasia/patología , Células Neoplásicas Circulantes , Unión Proteica , Receptores de Superficie Celular
15.
Semin Cell Dev Biol ; 67: 141-152, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-27641825

RESUMEN

Human tissues are remarkably adaptable and robust, harboring the collective ability to detect and respond to external stresses while maintaining tissue integrity. Following injury, many tissues have the capacity to repair the damage - and restore form and function - by deploying cellular and molecular mechanisms reminiscent of developmental programs. Indeed, it is increasingly clear that cancer and chronic conditions that develop with age arise as a result of cells and tissues re-implementing and deregulating a selection of developmental programs. Therefore, understanding the fundamental molecular mechanisms that drive cell and tissue responses is a necessity when designing therapies to treat human conditions. Extracellular matrix stiffness synergizes with chemical cues to drive single cell and collective cell behavior in culture and acts to establish and maintain tissue homeostasis in the body. This review will highlight recent advances that elucidate the impact of matrix mechanics on cell behavior and fate across these length scales during times of homeostasis and in disease states.


Asunto(s)
Neoplasias de la Mama/genética , Cadherinas/genética , Proteínas Contráctiles/genética , Células Epiteliales/metabolismo , Glándulas Mamarias Humanas/metabolismo , Mecanotransducción Celular , Adaptación Fisiológica , Animales , Fenómenos Biomecánicos , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Cadherinas/metabolismo , Proteínas Contráctiles/metabolismo , Citoesqueleto/metabolismo , Citoesqueleto/ultraestructura , Células Epiteliales/citología , Transición Epitelial-Mesenquimal , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Adhesiones Focales/metabolismo , Adhesiones Focales/ultraestructura , Regulación de la Expresión Génica , Homeostasis , Humanos , Glándulas Mamarias Humanas/citología , Estrés Mecánico
16.
J Cell Sci ; 130(1): 71-82, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28043968

RESUMEN

All cells sense and integrate mechanical and biochemical cues from their environment to orchestrate organismal development and maintain tissue homeostasis. Mechanotransduction is the evolutionarily conserved process whereby mechanical force is translated into biochemical signals that can influence cell differentiation, survival, proliferation and migration to change tissue behavior. Not surprisingly, disease develops if these mechanical cues are abnormal or are misinterpreted by the cells - for example, when interstitial pressure or compression force aberrantly increases, or the extracellular matrix (ECM) abnormally stiffens. Disease might also develop if the ability of cells to regulate their contractility becomes corrupted. Consistently, disease states, such as cardiovascular disease, fibrosis and cancer, are characterized by dramatic changes in cell and tissue mechanics, and dysregulation of forces at the cell and tissue level can activate mechanosignaling to compromise tissue integrity and function, and promote disease progression. In this Commentary, we discuss the impact of cell and tissue mechanics on tissue homeostasis and disease, focusing on their role in brain development, homeostasis and neural degeneration, as well as in brain cancer.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Matriz Extracelular/metabolismo , Homeostasis , Animales , Fenómenos Biomecánicos , Humanos , Mecanotransducción Celular , Microambiente Tumoral
17.
Phys Biol ; 15(2): 026002, 2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29116056

RESUMEN

Thermal fluctuations in cell membranes manifest as an excess area ([Formula: see text]) which governs a multitude of physical process at the sub-micron scale. We present a theoretical framework, based on an in silico tether pulling method, which may be used to reliably estimate [Formula: see text] in live cells. We perform our simulations in two different thermodynamic ensembles: (i) the constant projected area and (ii) the constant frame tension ensembles and show the equivalence of our results in the two. The tether forces estimated from our simulations compare well with our experimental measurements for tethers extracted from ruptured GUVs and HeLa cells. We demonstrate the significance and validity of our method by showing that all our calculations performed in the initial tether formation regime (i.e. when the length of the tether is comparable to its radius) along with experiments of tether extraction in 15 different cell types collapse onto two unified scaling relationships mapping tether force, tether radius, bending stiffness κ, and membrane tension σ. We show that [Formula: see text] is an important determinant of the radius of the extracted tether, which is equal to the characteristic length [Formula: see text] for [Formula: see text], and is equal to [Formula: see text] for [Formula: see text]. We also find that the estimated excess area follows a linear scaling behavior that only depends on the true value of [Formula: see text] for the membrane, based on which we propose a self-consistent technique to estimate the range of excess membrane areas in a cell.


Asunto(s)
Membrana Celular/fisiología , Simulación por Computador , Modelos Biológicos , Termodinámica
19.
Cancer Metastasis Rev ; 35(4): 655-667, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27914000

RESUMEN

The extracellular matrix (ECM) is a guiding force that regulates various developmental stages of the breast. In addition to providing structural support for the cells, it mediates epithelial-stromal communication and provides cues for cell survival, proliferation, and differentiation. Perturbations in ECM architecture profoundly influence breast tumor progression and metastasis. Understanding how a dysregulated ECM can facilitate malignant transformation is crucial to designing treatments to effectively target the tumor microenvironment. Here, we address the contribution of ECM mechanics to breast cancer progression, metastasis, and treatment resistance and discuss potential therapeutic strategies targeting the ECM.


Asunto(s)
Neoplasias de la Mama/patología , Transformación Celular Neoplásica/patología , Matriz Extracelular/patología , Animales , Humanos , Neoplasias Mamarias Experimentales/patología , Metástasis de la Neoplasia
20.
Hepatology ; 64(1): 261-75, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26755329

RESUMEN

UNLABELLED: Matrix rigidity has important effects on cell behavior and is increased during liver fibrosis; however, its effect on primary hepatocyte function is unknown. We hypothesized that increased matrix rigidity in fibrotic livers would activate mechanotransduction in hepatocytes and lead to inhibition of liver-specific functions. To determine the physiologically relevant ranges of matrix stiffness at the cellular level, we performed detailed atomic force microscopy analysis across liver lobules from normal and fibrotic livers. We determined that normal liver matrix stiffness was around 150 Pa and increased to 1-6 kPa in areas near fibrillar collagen deposition in fibrotic livers. In vitro culture of primary hepatocytes on collagen matrix of tunable rigidity demonstrated that fibrotic levels of matrix stiffness had profound effects on cytoskeletal tension and significantly inhibited hepatocyte-specific functions. Normal liver stiffness maintained functional gene regulation by hepatocyte nuclear factor 4 alpha (HNF4α), whereas fibrotic matrix stiffness inhibited the HNF4α transcriptional network. Fibrotic levels of matrix stiffness activated mechanotransduction in primary hepatocytes through focal adhesion kinase. In addition, blockade of the Rho/Rho-associated protein kinase pathway rescued HNF4α expression from hepatocytes cultured on stiff matrix. CONCLUSION: Fibrotic levels of matrix stiffness significantly inhibit hepatocyte-specific functions in part by inhibiting the HNF4α transcriptional network mediated through the Rho/Rho-associated protein kinase pathway. Increased appreciation of the role of matrix rigidity in modulating hepatocyte function will advance our understanding of the mechanisms of hepatocyte dysfunction in liver cirrhosis and spur development of novel treatments for chronic liver disease. (Hepatology 2016;64:261-275).


Asunto(s)
Matriz Extracelular/fisiología , Factor Nuclear 4 del Hepatocito/metabolismo , Hepatocitos/fisiología , Animales , Células Cultivadas , Citoesqueleto/fisiología , Expresión Génica , Cirrosis Hepática/metabolismo , Mecanotransducción Celular , Ratones Endogámicos C57BL , Microscopía de Fuerza Atómica , Quinasas Asociadas a rho/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA