Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 131
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 630(8018): 968-975, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38867043

RESUMEN

Obesity is a leading risk factor for progression and metastasis of many cancers1,2, yet can in some cases enhance survival3-5 and responses to immune checkpoint blockade therapies, including anti-PD-1, which targets PD-1 (encoded by PDCD1), an inhibitory receptor expressed on immune cells6-8. Although obesity promotes chronic inflammation, the role of the immune system in the obesity-cancer connection and immunotherapy remains unclear. It has been shown that in addition to T cells, macrophages can express PD-19-12. Here we found that obesity selectively induced PD-1 expression on tumour-associated macrophages (TAMs). Type I inflammatory cytokines and molecules linked to obesity, including interferon-γ, tumour necrosis factor, leptin, insulin and palmitate, induced macrophage PD-1 expression in an mTORC1- and glycolysis-dependent manner. PD-1 then provided negative feedback to TAMs that suppressed glycolysis, phagocytosis and T cell stimulatory potential. Conversely, PD-1 blockade increased the level of macrophage glycolysis, which was essential for PD-1 inhibition to augment TAM expression of CD86 and major histocompatibility complex I and II molecules and ability to activate T cells. Myeloid-specific PD-1 deficiency slowed tumour growth, enhanced TAM glycolysis and antigen-presentation capability, and led to increased CD8+ T cell activity with a reduced level of markers of exhaustion. These findings show that obesity-associated metabolic signalling and inflammatory cues cause TAMs to induce PD-1 expression, which then drives a TAM-specific feedback mechanism that impairs tumour immune surveillance. This may contribute to increased cancer risk yet improved response to PD-1 immunotherapy in obesity.


Asunto(s)
Neoplasias , Obesidad , Receptor de Muerte Celular Programada 1 , Macrófagos Asociados a Tumores , Animales , Femenino , Humanos , Masculino , Ratones , Presentación de Antígeno/efectos de los fármacos , Antígeno B7-2/antagonistas & inhibidores , Antígeno B7-2/inmunología , Antígeno B7-2/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Glucólisis/efectos de los fármacos , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Activación de Linfocitos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Ratones Endogámicos C57BL , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología , Obesidad/inmunología , Obesidad/metabolismo , Fagocitosis/efectos de los fármacos , Receptor de Muerte Celular Programada 1/metabolismo , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/metabolismo , Macrófagos Asociados a Tumores/efectos de los fármacos
2.
Physiol Rev ; 100(2): 695-724, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31751165

RESUMEN

Physical stimuli are essential for the function of eukaryotic cells, and changes in physical signals are important elements in normal tissue development as well as in disease initiation and progression. The complexity of physical stimuli and the cellular signals they initiate are as complex as those triggered by chemical signals. One of the most important, and the focus of this review, is the effect of substrate mechanical properties on cell structure and function. The past decade has produced a nearly exponentially increasing number of mechanobiological studies to define how substrate stiffness alters cell biology using both purified systems and intact tissues. Here we attempt to identify common features of mechanosensing in different systems while also highlighting the numerous informative exceptions to what in early studies appeared to be simple rules by which cells respond to mechanical stresses.


Asunto(s)
Microambiente Celular , Mecanotransducción Celular , Animales , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Forma de la Célula , Elasticidad , Humanos , Viscosidad
3.
J Cell Sci ; 136(14)2023 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-37358264

RESUMEN

Cancer-associated fibroblasts (CAFs) have distinct roles within the tumor microenvironment, which can impact the mode and efficacy of tumor cell migration. CAFs are known to increase invasion of less-aggressive breast cancer cells through matrix remodeling and leader-follower dynamics. Here, we demonstrate that CAFs communicate with breast cancer cells through the formation of contact-dependent tunneling nanotubes (TNTs), which allow for the exchange of cargo between cell types. CAF mitochondria are an integral cargo component and are sufficient to increase the 3D migration of cancer cells. This cargo transfer results in an increase in mitochondrial ATP production in cancer cells, whereas it has a negligible impact on glycolytic ATP production. Manually increasing mitochondrial oxidative phosphorylation (OXPHOS) by providing extra substrates for OXPHOS fails to enhance cancer cell migration unless glycolysis is maintained at a constant level. Together, these data indicate that tumor-stromal cell crosstalk via TNTs and the associated metabolic symbiosis is a finely controlled mechanism by which tumor cells co-opt their microenvironment to promote cancer progression and may become a potential therapeutic target.


Asunto(s)
Neoplasias de la Mama , Fibroblastos Asociados al Cáncer , Humanos , Femenino , Neoplasias de la Mama/patología , Fibroblastos Asociados al Cáncer/metabolismo , Línea Celular Tumoral , Mitocondrias/metabolismo , Adenosina Trifosfato/metabolismo , Fibroblastos/metabolismo , Microambiente Tumoral
5.
Proc Natl Acad Sci U S A ; 119(18): e2114672119, 2022 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-35471912

RESUMEN

Altered tissue mechanics and metabolism have gained significant attention as drivers of tumorigenesis, and mechanoresponsive metabolism has been implicated in migration and metastasis. However, heterogeneity in cell populations makes it difficult to link changes in behavior with metabolism, as individual cell behaviors are not necessarily reflected in population-based measurements. As such, the impact of increased collagen deposition, a tumor-associated collagen signature, on metabolism remains ambiguous. Here, we utilize a wide range of collagen densities to alter migration ability and study the bioenergetics of individual cells over time. Sorting cells based on their level of motility revealed energetics are a function of collagen density only for highly motile cells, not the entire population or cells with low motility. Changes in migration with increasing collagen density were correlated with cellular energetics, where matrix conditions most permissive to migration required less energy usage during movement and migrated more efficiently. These findings reveal a link between matrix mechanics, migratory phenotype, and bioenergetics and suggest that energetic costs are determined by the extracellular matrix and influence cell motility.


Asunto(s)
Colágeno , Neoplasias , Movimiento Celular/fisiología , Transformación Celular Neoplásica/metabolismo , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Humanos , Neoplasias/metabolismo
6.
Biol Cell ; 115(8): e2200104, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37224184

RESUMEN

Although it is well-known that cancer-associated fibroblasts (CAFs) play a key role in regulating tumor progression, the effects of mechanical tissue changes on CAFs are understudied. Myofibroblastic CAFs (myCAFs), in particular, are known to alter tumor matrix architecture and composition, heavily influencing the mechanical forces in the tumor microenvironment (TME), but much less is known about how these mechanical changes initiate and maintain the myCAF phenotype. Additionally, recent studies have pointed to the existence of CAFs in circulating tumor cell clusters, indicating that CAFs may be subject to mechanical forces beyond the primary TME. Due to their pivotal role in cancer progression, targeting CAF mechanical regulation may provide therapeutic benefit. Here, we will discuss current knowledge and summarize existing gaps in how CAFs regulate and are regulated by matrix mechanics, including through stiffness, solid and fluid stresses, and fluid shear stress.


Asunto(s)
Fibroblastos Asociados al Cáncer , Neoplasias , Humanos , Fibroblastos Asociados al Cáncer/patología , Fibroblastos/patología , Neoplasias/patología , Microambiente Tumoral
7.
Breast Cancer Res ; 25(1): 102, 2023 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-37649089

RESUMEN

BACKGROUND: Intratumor heterogeneity is a well-established hallmark of cancer that impedes cancer research, diagnosis, and treatment. Previously, we phenotypically sorted human breast cancer cells based on migratory potential. When injected into mice, highly migratory cells were weakly metastatic and weakly migratory cells were highly metastatic. The purpose of this study was to determine whether these weakly and highly migratory cells interact with each other in vitro or in vivo. METHODS: To assess the relationship between heterogeneity in cancer cell migration and metastatic fitness, MDA-MB-231 and SUM159PT triple negative breast cancer cells were phenotypically sorted into highly migratory and weakly migratory subpopulations and assayed separately and in a 1:1 mixture in vitro and in vivo for metastatic behaviors. Unpaired, two-tailed Student's t-tests, Mann-Whitney tests, ordinary, one-way ANOVAs, and Kruskal-Wallis H tests were performed as appropriate with p < 0.05 as the cutoff for statistical significance. RESULTS: When highly and weakly migratory cells are co-seeded in mixed spheroids, the weakly migratory cells migrated farther than weakly migratory only spheroids. In mixed spheroids, leader-follower behavior occurred with highly migratory cells leading the weakly migratory cells in migration strands. When cell suspensions of highly migratory, weakly migratory, or a 1:1 mixture of both subpopulations were injected orthotopically into mice, both the mixed cell suspensions and weakly migratory cells showed significant distal metastasis, but the highly migratory cells did not metastasize significantly to any location. Notably, significantly more distal metastasis was observed in mice injected with the 1:1 mixture compared to either subpopulation alone. CONCLUSIONS: This study suggests that weakly migratory cells interact with highly migratory cells in a commensal fashion resulting in increased migration and metastasis. Together, these findings indicate that cancer cell subpopulation migration ability does not correlate with metastatic potential and that cooperation between highly migratory and weakly migratory subpopulations can enhance overall metastatic fitness.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Animales , Ratones , Suspensiones , Simbiosis , Movimiento Celular , Bioensayo
8.
BMC Biol ; 20(1): 61, 2022 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-35260156

RESUMEN

BACKGROUND: T cell activation is a mechanical process as much as it is a biochemical process. In this study, we used a cone-and-plate viscometer system to treat Jurkat and primary human T cells with fluid shear stress (FSS) to enhance the activation of the T cells through mechanical means. RESULTS: The FSS treatment of T cells in combination with soluble and bead-bound CD3/CD28 antibodies increased the activation of signaling proteins essential for T cell activation, such as zeta-chain-associated protein kinase-70 (ZAP70), nuclear factor of activated T cells (NFAT), nuclear factor kappa B (NF-κB), and AP-1 (activator protein 1). The FSS treatment also enhanced the expression of the cytokines tumor necrosis factor alpha (TNF-α), interleukin 2 (IL-2), and interferon gamma (IFN-γ), which are necessary for sustained T cell activation and function. The enhanced activation of T cells by FSS was calcium dependent. The calcium signaling was controlled by the mechanosensitive ion channel Piezo1, as GsMTx-4 and Piezo1 knockout reduced ZAP70 phosphorylation by FSS. CONCLUSIONS: These results demonstrate an intriguing new dynamic to T cell activation, as the circulatory system consists of different magnitudes of FSS and could have a proinflammatory role in T cell function. The results also identify a potential pathophysiological relationship between T cell activation and FSS, as hypertension is a disease characterized by abnormal blood flow and is correlated with multiple autoimmune diseases.


Asunto(s)
Señalización del Calcio , Canales Iónicos , Humanos , Canales Iónicos/metabolismo , Fosforilación , Estrés Mecánico , Factor de Necrosis Tumoral alfa/metabolismo
9.
J Cell Sci ; 133(18)2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32878941

RESUMEN

Intimal stiffening upregulates endothelial cell contractility, disrupting barrier integrity; however, intimal stiffening is non-uniform. The impact of local changes in intimal stiffness on proximal and distal cell-cell interactions is unknown. To investigate the range at which matrix stiffness heterogeneities impact neighboring endothelial cells within a monolayer, we built a micropillar system with adjacent regions of stiff and compliant matrix. The stiffness interface results in an oscillatory pattern of neutrophil transendothelial migration, symmetrical about the interface and well-fit by a sinusoid function. 'Peaks' of the sinusoid were found to have increased cellular contractility and decreased barrier function relative to 'troughs' of the sinusoid. Pharmacological modulation of contractility was observed to break symmetry, altering the amplitude and wavelength of the sinusoid, indicating that contractility may regulate this effect. This work illuminates a novel biophysical phenomenon of the role of stiffness-mediated cell-matrix interactions on cell-cell interactions at a distance. Additionally, it provides insight into the range at which intimal matrix stiffness heterogeneities will impact endothelial barrier function and potentially contribute to atherogenesis.


Asunto(s)
Aterosclerosis , Células Endoteliales , Comunicación Celular , Movimiento Celular , Matriz Extracelular , Humanos
10.
J Cell Sci ; 133(1)2020 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-31822629

RESUMEN

Abnormal tensional cellular homeostasis is now considered a hallmark of cancer. Despite this, the origin of this abnormality remains unclear. In this work, we investigated the role of tissue transglutaminase 2 (TG2, also known as TGM2), a protein associated with poor prognosis and increased metastatic potential, and its relationship to the EGF receptor in the regulation of the mechanical state of tumor cells. Remarkably, we observed a TG2-mediated modulation of focal adhesion composition as well as stiffness-induced FAK activation, which was linked with a distinctive increase in cell contractility, in experiments using both pharmacological and shRNA-based approaches. Additionally, the increased contractility could be reproduced in non-malignant cells upon TG2 expression. Moreover, the increased cell contractility mediated by TG2 was largely due to the loss of EGFR-mediated inhibition of cell contractility. These findings establish intracellular TG2 as a regulator of cellular tensional homeostasis and suggest the existence of signaling switches that control the contribution of growth factor receptors in determining the mechanical state of a cell.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Transglutaminasas/metabolismo , Fenómenos Biomecánicos , Línea Celular Tumoral , Forma de la Célula/fisiología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Receptores ErbB/metabolismo , Adhesiones Focales/metabolismo , Homeostasis , Humanos , Proteína Glutamina Gamma Glutamiltransferasa 2 , Transducción de Señal
11.
Soft Matter ; 18(44): 8504-8513, 2022 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-36325938

RESUMEN

Diabetes is associated with increased risk of breast cancer and worse prognoses for cancer patients. Hyperglycemia can result in increased glycation, the process wherein crosslinkages are formed between sugars and extracellular matrix (ECM) proteins through the formation of advanced glycation endproducts (AGEs). Although accumulation of AGEs occurs naturally in vivo over time, it is greatly accelerated by the hyperglycemic environment of diabetic patients. AGE accumulation has been linked to stiffening-related diseases such as hypertension, cancer metastasis, and neurodegenerative disorders. In response, several AGE-inhibiting and AGE-breaking drugs have received significant attention for their ability to reduce AGE accumulation. The resulting effects of these drugs on cell behavior is not well understood. In this study, we measured cancer cell migration in glycated collagen with and without the AGE-breaking drug alagebrium chloride (ALT711) to investigate the drug's ability to disrupt ECM crosslinks and reduce tumor cell spreading, contractility, and migration. The mechanical properties and chemical composition of collagen glycated with increasing concentrations of glucose with and without ALT711 treatment were measured. Increasing glucose concentration resulted in increased AGE accumulation and matrix stiffness as well as increased cancer cell contractility, elongation, and migration. Treatment with ALT711 significantly lowered AGE accumulation within the collagen, decreased collagen stiffness, and reduced cell migration. These findings suggest that while hyperglycemia can increase collagen matrix stiffness, resulting in increased breast cancer cell migration, an AGE-breaker can reverse this phenotype and may be a viable treatment option for reducing cancer cell migration due to glycation.


Asunto(s)
Hiperglucemia , Neoplasias , Humanos , Productos Finales de Glicación Avanzada/metabolismo , Movimiento Celular , Colágeno/metabolismo , Glucosa , Neoplasias/tratamiento farmacológico
12.
Proc Natl Acad Sci U S A ; 116(16): 7867-7872, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30923113

RESUMEN

The ability of primary tumor cells to invade into adjacent tissues, followed by the formation of local or distant metastasis, is a lethal hallmark of cancer. Recently, locomoting clusters of tumor cells have been identified in numerous cancers and associated with increased invasiveness and metastatic potential. However, how the collective behaviors of cancer cells are coordinated and their contribution to cancer invasion remain unclear. Here we show that collective invasion of breast cancer cells is regulated by the energetic statuses of leader and follower cells. Using a combination of in vitro spheroid and ex vivo organoid invasion models, we found that cancer cells dynamically rearrange leader and follower positions during collective invasion. Cancer cells invade cooperatively in denser collagen matrices by accelerating leader-follower switching thus decreasing leader cell lifetime. Leader cells exhibit higher glucose uptake than follower cells. Moreover, their energy levels, as revealed by the intracellular ATP/ADP ratio, must exceed a threshold to invade. Forward invasion of the leader cell gradually depletes its available energy, eventually leading to leader-follower transition. Our computational model based on intracellular energy homeostasis successfully recapitulated the dependence of leader cell lifetime on collagen density. Experiments further supported model predictions that decreasing the cellular energy level by glucose starvation decreases leader cell lifetime whereas increasing the cellular energy level by AMP-activated kinase (AMPK) activation does the opposite. These findings highlight coordinated invasion and its metabolic regulation as potential therapeutic targets of cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/fisiopatología , Movimiento Celular/fisiología , Metabolismo Energético/fisiología , Invasividad Neoplásica/fisiopatología , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Línea Celular Tumoral , Femenino , Glucosa/metabolismo , Humanos , Espacio Intracelular/metabolismo
13.
Biophys J ; 120(9): 1705-1717, 2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33705759

RESUMEN

The microenvironment provides both active and passive mechanical cues that regulate cell morphology, adhesion, migration, and metabolism. Although the cellular response to those mechanical cues often requires energy-intensive actin cytoskeletal remodeling and actomyosin contractility, it remains unclear how cells dynamically adapt their metabolic activity to altered mechanical cues to support migration. Here, we investigated the changes in cellular metabolic activity in response to different two-dimensional and three-dimensional microenvironmental conditions and how these changes relate to cytoskeletal activity and migration. Utilizing collagen micropatterning on polyacrylamide gels, intracellular energy levels and oxidative phosphorylation were found to be correlated with cell elongation and spreading and necessary for membrane ruffling. To determine whether this relationship holds in more physiological three-dimensional matrices, collagen matrices were used to show that intracellular energy state was also correlated with protrusive activity and increased with matrix density. Pharmacological inhibition of oxidative phosphorylation revealed that cancer cells rely on oxidative phosphorylation to meet the elevated energy requirements for protrusive activity and migration in denser matrices. Together, these findings suggest that mechanical regulation of cytoskeletal activity during spreading and migration by the physical microenvironment is driven by an altered metabolic profile.


Asunto(s)
Citoesqueleto , Matriz Extracelular , Actomiosina , Movimiento Celular , Colágeno
14.
J Cell Sci ; 131(12)2018 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-29748381

RESUMEN

Force generation within cells, mediated by motor proteins along cytoskeletal networks, maintains the function of multicellular structures during homeostasis and when generating collective forces. Here, we describe the use of chromatin dynamics to detect cellular force propagation [a technique termed SINK (sensors from intranuclear kinetics)] and investigate the force response of cells to disruption of the monolayer and changes in substrate stiffness. We find that chromatin dynamics change in a substrate stiffness-dependent manner within epithelial monolayers. We also investigate point defects within monolayers to map the impact on the strain field of a heterogeneous monolayer. We find that cell monolayers behave as a colloidal assembly rather than as a continuum since the data fit an exponential decay; the lateral characteristic length of recovery from the mechanical defect is ∼50 µm for cells with a 10 µm spacing. At distances greater than this characteristic length, cells behave similarly to those in a fully intact monolayer. This work demonstrates the power of SINK to investigate diseases including cancer and atherosclerosis that result from single cells or heterogeneities in monolayers.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Citoesqueleto/fisiología , Células Epiteliales/citología , Fenómenos Biomecánicos , Cromatina/fisiología , Humanos , Reología/métodos
15.
FASEB J ; 33(1): 1199-1208, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30102569

RESUMEN

Tumor vasculature is known to be more permeable than the vasculature found in healthy tissue, which in turn can lead to a more aggressive tumor phenotype and impair drug delivery into tumors. While the stiffening of the stroma surrounding solid tumors has been reported to increase vascular permeability, the mechanism of this process remains unclear. Here, we utilize an in vitro model of tumor stiffening, ex ovo culture, and a mouse model to investigate the molecular mechanism by which matrix stiffening alters endothelial barrier function. Our data indicate that the increased endothelial permeability caused by heightened matrix stiffness can be prevented by pharmaceutical inhibition of focal adhesion kinase (FAK) both in vitro and ex ovo. Matrix stiffness-mediated FAK activation determines Src localization to cell-cell junctions, which then induces increased vascular endothelial cadherin phosphorylation both in vitro and in vivo. Endothelial cells in stiff tumors have more activated Src and higher levels of phosphorylated vascular endothelial cadherin at adherens junctions compared to endothelial cells in more compliant tumors. Altogether, our data indicate that matrix stiffness regulates endothelial barrier integrity through FAK activity, providing one mechanism by which extracellular matrix stiffness regulates endothelial barrier function. Additionally, our work also provides further evidence that FAK is a promising potential target for cancer therapy because FAK plays a critical role in the regulation of endothelial barrier integrity.-Wang, W., Lollis, E. M., Bordeleau, F., Reinhart-King, C. A. Matrix stiffness regulates vascular integrity through focal adhesion kinase activity.


Asunto(s)
Endotelio Vascular/metabolismo , Matriz Extracelular/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Uniones Adherentes/metabolismo , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Permeabilidad Capilar , Embrión de Pollo , Endotelio Vascular/citología , Activación Enzimática , Matriz Extracelular/enzimología , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Transgénicos , Fosforilación , Transporte de Proteínas , Tirosina/metabolismo , Familia-src Quinasas/metabolismo
16.
Curr Top Membr ; 86: 255-278, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33837695

RESUMEN

Microvesicles are small, membrane-bound vesicles that are shed from the plasma membrane of cells into the extracellular space. Microvesicles contain a variety of cargo not typically thought to be released from cells, including receptor tyrosine kinases, cytosolic signaling proteins, and microRNAs, which are transferred from donor cells to recipient cells. The transfer of microvesicle cargo can result in the transformation of recipient cells thereby supporting disease progression, including modified fibroblast metabolism, epithelial cell contractility, vascular remodeling, and immune cell inflammatory signaling. Additionally, microvesicles are believed to play prominent roles in cell-cell communication and disease progression as they are detected at elevated concentrations in diseased tissues. As microvesicle uptake by recipient cells can modulate cell function to promote disease progression, understanding the mechanisms and mechanosensitivity of microvesicle release, internalization, and the resulting signaling is crucial to fully comprehend their functions in disease. Here, we review recent advances in the understanding of actomyosin-regulated microvesicle biogenesis, microvesicle uptake via pinocytosis, and the resulting cellular transformation. We discuss the effects of altered cell contractility, mode of cell migration, and extracellular matrix compliance on microvesicle signaling, with direct implications in disease progression and identifying future therapeutic targets.


Asunto(s)
Micropartículas Derivadas de Células , Transporte Biológico , Biofisica , Membrana Celular/metabolismo , Micropartículas Derivadas de Células/metabolismo , Matriz Extracelular/metabolismo
17.
Proc Natl Acad Sci U S A ; 114(3): 492-497, 2017 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-28034921

RESUMEN

Tumor microvasculature tends to be malformed, more permeable, and more tortuous than vessels in healthy tissue, effects that have been largely attributed to up-regulated VEGF expression. However, tumor tissue tends to stiffen during solid tumor progression, and tissue stiffness is known to alter cell behaviors including proliferation, migration, and cell-cell adhesion, which are all requisite for angiogenesis. Using in vitro, in vivo, and ex ovo models, we investigated the effects of matrix stiffness on vessel growth and integrity during angiogenesis. Our data indicate that angiogenic outgrowth, invasion, and neovessel branching increase with matrix cross-linking. These effects are caused by increased matrix stiffness independent of matrix density, because increased matrix density results in decreased angiogenesis. Notably, matrix stiffness up-regulates matrix metalloproteinase (MMP) activity, and inhibiting MMPs significantly reduces angiogenic outgrowth in stiffer cross-linked gels. To investigate the functional significance of altered endothelial cell behavior in response to matrix stiffness, we measured endothelial cell barrier function on substrates mimicking the stiffness of healthy and tumor tissue. Our data indicate that barrier function is impaired and the localization of vascular endothelial cadherin is altered as function of matrix stiffness. These results demonstrate that matrix stiffness, separately from matrix density, can alter vascular growth and integrity, mimicking the changes that exist in tumor vasculature. These data suggest that therapeutically targeting tumor stiffness or the endothelial cell response to tumor stiffening may help restore vessel structure, minimize metastasis, and aid in drug delivery.


Asunto(s)
Matriz Extracelular/fisiología , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/fisiopatología , Microvasos/fisiopatología , Animales , Fenómenos Biomecánicos , Bovinos , Células Cultivadas , Embrión de Pollo , Colágeno/metabolismo , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Neoplasias Mamarias Experimentales/patología , Metaloproteinasas de la Matriz/metabolismo , Ratones , Microvasos/patología , Invasividad Neoplásica/patología , Invasividad Neoplásica/fisiopatología , Neovascularización Patológica/patología , Neovascularización Patológica/fisiopatología , Fenotipo , Microambiente Tumoral/fisiología , Rigidez Vascular/fisiología
18.
Biophys J ; 117(9): 1692-1701, 2019 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-31623884

RESUMEN

During metastasis, cancer cells navigate through a spatially heterogeneous extracellular matrix (ECM). Physical properties of ECM, including the degree of confinement, influence cell migration behavior. Here, utilizing in vitro three-dimensional collagen microtracks, we demonstrate that cell-ECM interactions, specifically the degree of spatial confinement, regulate migratory behavior. We found that cells migrate faster when they are fully confined, contacting all four walls (top, bottom, and two sides) of a collagen microtrack, compared with cells that are partially confined, contacting less than four walls. When fully confined, cells exhibit fewer but larger vinculin-containing adhesions and create greater strains in the surrounding matrix directed toward the cell body. In contrast, partially confined cells develop a more elongated morphology with smaller but significantly more vinculin-containing adhesions and displace the surrounding matrix less than fully confined cells. The resulting effect of increasing cell contractility via Rho activation is dependent on the number of walls with which the cell is in contact. Although matrix strains increase in both fully and partially confined cells, cells that are partially confined increase speed, whereas those in full confinement decrease speed. Together, these results suggest that the degree of cell-ECM contact during confined migration is a key determinant of speed, morphology, and cell-generated substrate strains during motility, and these factors may work in tandem to facilitate metastatic cell migration.


Asunto(s)
Movimiento Celular , Matriz Extracelular/metabolismo , Línea Celular Tumoral , Tamaño de la Célula , Uniones Célula-Matriz/metabolismo , Activación Enzimática , Adhesiones Focales/metabolismo , Humanos , Vinculina/metabolismo , Proteínas de Unión al GTP rho/metabolismo
19.
Am J Physiol Cell Physiol ; 317(1): C82-C92, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31017799

RESUMEN

Extracellular vesicles released by cancer cells have recently been implicated in the differentiation of stromal cells to their activated, cancer-supporting states. Microvesicles, a subset of extracellular vesicles released from the plasma membrane of cancer cells, contain biologically active cargo, including DNA, mRNA, and miRNA, which are transferred to recipient cells and induce a phenotypic change in behavior. While it is known that microvesicles can alter recipient cell phenotype, little is known about how the physical properties of the tumor microenvironment affect fibroblast response to microvesicles. Here, we utilized cancer cell-derived microvesicles and synthetic substrates designed to mimic the stiffness of the tumor and tumor stroma to investigate the effects of microvesicles on fibroblast phenotype as a function of the mechanical properties of the microenvironment. We show that microvesicles released by highly malignant breast cancer cells cause an increase in fibroblast spreading, α-smooth muscle actin expression, proliferation, cell-generated traction force, and collagen gel compaction. Notably, our data indicate that these phenotypic changes occur only on stiff matrices mimicking the stiffness of the tumor periphery and are dependent on the cell type from which the microvesicles are shed. Overall, these results show that the effects of cancer cell-derived microvesicles on fibroblast activation are regulated by the physical properties of the microenvironment, and these data suggest that microvesicles may have a more robust effect on fibroblasts located at the tumor periphery to influence cancer progression.


Asunto(s)
Neoplasias de la Mama/patología , Fibroblastos Asociados al Cáncer/patología , Micropartículas Derivadas de Células/patología , Matriz Extracelular/patología , Mecanotransducción Celular , Comunicación Paracrina , Actinas/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Proliferación Celular , Micropartículas Derivadas de Células/metabolismo , Módulo de Elasticidad , Matriz Extracelular/metabolismo , Femenino , Humanos , Células MCF-7 , Ratones , Células 3T3 NIH , Fenotipo , Microambiente Tumoral
20.
J Biomech Eng ; 140(7)2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29560498

RESUMEN

Aerobic exercise helps to maintain cardiovascular health in part by mitigating age-induced arterial stiffening. However, the long-term effects of exercise regimens on aortic stiffness remain unknown, especially in the intimal extracellular matrix layer known as the subendothelial matrix. To examine how the stiffness of the subendothelial matrix changes following exercise cessation, mice were exposed to an 8 week swimming regimen followed by an 8 week sedentary rest period. Whole vessel and subendothelial matrix stiffness were measured after both the exercise and rest periods. After swimming, whole vessel and subendothelial matrix stiffness decreased, and after 8 weeks of rest, these values returned to baseline. Within the same time frame, the collagen content in the intima layer and the presence of advanced glycation end products (AGEs) in the whole vessel were also affected by the exercise and the rest periods. Overall, our data indicate that consistent exercise is necessary for maintaining compliance in the subendothelial matrix.


Asunto(s)
Endotelio Vascular/metabolismo , Fenómenos Mecánicos , Condicionamiento Físico Animal , Animales , Aorta/citología , Aorta/fisiología , Fenómenos Biomecánicos , Colágeno/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis de la Onda del Pulso , Descanso , Volumen Sistólico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA