Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 36(5): e22278, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35436025

RESUMEN

Fluid shear stress is an important mediator of vascular permeability, yet the molecular mechanisms underlying the effect of shear on the blood-brain barrier (BBB) have yet to be clarified in cerebral vasculature despite its importance for brain homeostasis. The goal of this study is to probe components of shear mechanotransduction within the BBB to gain a better understanding of pathologies associated with changes in cerebral perfusion including ischemic stroke. Interrogating the effects of shear stress in vivo is complicated by the complexity of factors in the brain parenchyma and the difficulty associated with modulating blood flow regimes. The in vitro model used in this study is compatible with real-time measurement of barrier function using a transendothelial electrical resistance as well as immunocytochemistry and dextran permeability assays. These experiments reveal that there is a threshold level of shear stress required for barrier formation and that the composition of the extracellular matrix, specifically the presence of high molecular weight hyaluronan, dictates the flow response. Gene editing to modulate the expression of CD44, a mechanosensitive receptor for hyaluronan, demonstrates that the receptor is required for the endothelial response to shear stress. Manipulation of small GTPase activity reveals CD44 activates Rac1 while inhibiting RhoA activation. Additionally, adducin-γ localizes to tight junctions in response to shear stress and RhoA inhibition and is required to maintain the barrier. This study identifies specific components of the mechanosensing complex associated with the BBB response to fluid shear stress and, therefore, illuminates potential targets for barrier manipulation in vivo.


Asunto(s)
Barrera Hematoencefálica , Proteínas de Unión al GTP Monoméricas , Barrera Hematoencefálica/metabolismo , Permeabilidad Capilar/fisiología , Ácido Hialurónico/metabolismo , Mecanotransducción Celular , Proteínas de Unión al GTP Monoméricas/metabolismo
2.
Biochem J ; 479(17): 1825-1842, 2022 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-36094371

RESUMEN

Cell stiffness is an important characteristic of cells and their response to external stimuli. In this review, we survey methods used to measure cell stiffness, summarize stimuli that alter cell stiffness, and discuss signaling pathways and mechanisms that control cell stiffness. Several pathological states are characterized by changes in cell stiffness, suggesting this property can serve as a potential diagnostic marker or therapeutic target. Therefore, we consider the effect of cell stiffness on signaling and growth processes required for homeostasis and dysfunction in healthy and pathological states. Specifically, the composition and structure of the cell membrane and cytoskeleton are major determinants of cell stiffness, and studies have identified signaling pathways that affect cytoskeletal dynamics both directly and by altered gene expression. We present the results of studies interrogating the effects of biophysical and biochemical stimuli on the cytoskeleton and other cellular components and how these factors determine the stiffness of both individual cells and multicellular structures. Overall, these studies represent an intersection of the fields of polymer physics, protein biochemistry, and mechanics, and identify specific mechanisms involved in mediating cell stiffness that can serve as therapeutic targets.


Asunto(s)
Citoesqueleto , Transducción de Señal , Membrana Celular , Citoesqueleto/metabolismo , Microtúbulos
3.
Neurol Neurochir Pol ; 57(1): 14-25, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36810757

RESUMEN

The typical manifestation of coronavirus 2 (CoV-2) infection is a severe acute respiratory syndrome (SARS) accompanied by pneumonia (COVID-19). However, SARS-CoV-2 can also affect the brain, causing chronic neurological symptoms, variously known as long, post, post-acute, or persistent COVID-19 condition, and affecting up to 40% of patients. The symptoms (fatigue, dizziness, headache, sleep disorders, malaise, disturbances of memory and mood) usually are mild and resolve spontaneously. However, some patients develop acute and fatal complications, including stroke or encephalopathy. Damage to the brain vessels mediated by the coronavirus spike protein (S-protein) and overactive immune responses have been identified as leading causes of this condition. However, the molecular mechanism by which the virus affects the brain still needs to be fully delineated. In this review article, we focus on interactions between host molecules and S-protein as the mechanism allowing the transit of SARS-CoV-2 through the blood-brain barrier to reach the brain structures. In addition, we discuss the impact of S-protein mutations and the involvement of other cellular factors conditioning the pathophysiology of SARS-CoV-2 infection. Finally, we review current and future COVID-19 treatment options.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/metabolismo , Barrera Hematoencefálica/metabolismo , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/metabolismo , Tratamiento Farmacológico de COVID-19
4.
J Cell Physiol ; 237(10): 3872-3882, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35901247

RESUMEN

A variety of biophysical properties are known to regulate angiogenic sprouting, and in vitro systems can parse the individual effects of these factors in a controlled setting. Here, a three-dimensional brain microvascular model interrogates how variables including extracellular matrix composition, fluid shear stress, and radius of curvature affect angiogenic sprouting of cerebral endothelial cells. Tracking endothelial migration over several days reveals that application of fluid shear stress and enlarged vessel radius of curvature both attenuate sprouting. Computational modeling informed by oxygen consumption assays suggests that sprouting correlates to reduced oxygen concentration: both fluid shear stress and vessel geometry alter the local oxygen levels dictated by both ambient conditions and cellular respiration. Moreover, increasing cell density and consequently lowering the local oxygen levels yields significantly more sprouting. Further analysis reveals that the magnitude of oxygen concentration is not as important as its spatial concentration gradient: decreasing ambient oxygen concentration causes significantly less sprouting than applying an external oxygen gradient to the vessels. In contrast, barriergenesis is dictated by shear stress independent of local oxygen concentrations, suggesting that different mechanisms mediate angiogenesis and barrier formation and that angiogenic sprouting can occur without compromising the barrier. Overall, these results improve our understanding of how specific biophysical variables regulate the function and activation of cerebral vasculature, and identify spatial oxygen gradients as the driving factor of angiogenesis in the brain.


Asunto(s)
Células Endoteliales , Factor A de Crecimiento Endotelial Vascular , Encéfalo/metabolismo , Humanos , Neovascularización Patológica , Neovascularización Fisiológica , Oxígeno/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo
5.
J Neuroinflammation ; 19(1): 282, 2022 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-36434734

RESUMEN

BACKGROUND: Plasma gelsolin (pGSN) is an important part of the blood actin buffer that prevents negative consequences of possible F-actin deposition in the microcirculation and has various functions during host immune response. Recent reports reveal that severe COVID-19 correlates with reduced levels of pGSN. Therefore, using an in vitro system, we investigated whether pGSN could attenuate increased permeability of the blood-brain barrier (BBB) during its exposure to the portion of the SARS-CoV-2 spike protein containing the receptor binding domain (S1 subunit). MATERIALS AND METHODS: Two- and three-dimensional models of the human BBB were constructed using the human cerebral microvascular endothelial cell line hCMEC/D3 and exposed to physiologically relevant shear stress to mimic perfusion in the central nervous system (CNS). Trans-endothelial electrical resistance (TEER) as well as immunostaining and Western blotting of tight junction (TJ) proteins assessed barrier integrity in the presence of the SARS-CoV-2 spike protein and pGSN. The IncuCyte Live Imaging system evaluated the motility of the endothelial cells. Magnetic bead-based ELISA was used to determine cytokine secretion. Additionally, quantitative real-time PCR (qRT-PCR) revealed gene expression of proteins from signaling pathways that are associated with the immune response. RESULTS: pGSN reversed S1-induced BBB permeability in both 2D and 3D BBB models in the presence of shear stress. BBB models exposed to pGSN also exhibited attenuated pro-inflammatory signaling pathways (PI3K, AKT, MAPK, NF-κB), reduced cytokine secretion (IL-6, IL-8, TNF-α), and increased expression of proteins that form intercellular TJ (ZO-1, occludin, claudin-5). CONCLUSION: Due to its anti-inflammatory and protective effects on the brain endothelium, pGSN has the potential to be an alternative therapeutic target for patients with severe SARS-CoV-2 infection, especially those suffering neurological complications of COVID-19.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , Glicoproteína de la Espiga del Coronavirus , Barrera Hematoencefálica , Gelsolina/farmacología , Células Endoteliales , Permeabilidad , Proteínas de Uniones Estrechas , Citocinas
6.
Small ; 18(21): e2200883, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35451204

RESUMEN

Cellular mechanics encompass both mechanical properties that resist forces applied by the external environment and internally generated forces applied at the location of cell-cell and cell-matrix junctions. Here, the authors demonstrate that microindentation of cellular domes formed by cell monolayers that locally lift off the substrate provides insight into both aspects of cellular mechanics in multicellular structures. Using a modified Hertz contact equation, the force-displacement curves generated by a micro-tensiometer are used to measure an effective dome stiffness. The results indicate the domes are consistent with the Laplace-Young relationship for elastic membranes, regardless of biochemical modulation of the RhoA-ROCK signaling axis. In contrast, activating RhoA, and inhibiting ROCK both alter the relaxation dynamics of the domes deformed by the micro-tensiometer, revealing an approach to interrogate the role of RhoA-ROCK signaling in multicellular mechanics. A finite element model incorporating a Mooney-Rivlin hyperelastic constitutive equation to describe monolayer mechanics predicts effective stiffness values that are consistent with the micro-tensiometer measurements, verifying previous measurements of the response of cell monolayers to tension. Overall, these studies establish microindentation of fluid-filled domes as an avenue to investigate the contribution of cell-generated forces to the mechanics of multicellular structures.


Asunto(s)
Transducción de Señal
7.
Biotechnol Bioeng ; 118(7): 2411-2421, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33615435

RESUMEN

Previous in vitro studies interrogating the endothelial response to physiologically relevant flow regimes require specialized pumps to deliver time-dependent waveforms that imitate in vivo blood flow. The aim of this study is to create a low-cost and broadly adaptable approach to mimic physiological flow, and then use this system to characterize the effect of flow separation on velocity and shear stress profiles in a three-dimensional (3D) topology. The flow apparatus incorporates a programmable linear actuator that superposes oscillations on a constant mean flow driven by a peristaltic pump to emulate flow in the carotid artery. The flow is perfused through a 3D in vitro model of the blood-brain barrier designed to induce separated flow. Experimental flow patterns measured by microparticle image velocimetry and modeled by computational fluid dynamics reveal periodic changes in the instantaneous shear stress along the channel wall. Moreover, the time-dependent flow causes periodic flow separation zones, resulting in variable reattachment points during the cycle. The effects of these complex flow regimes are assessed by evaluating the integrity of the in vitro blood-brain barrier model. Permeability assays and immunostaining for proteins associated with tight junctions reveal barrier breakdown in the region of disturbed flow. In conclusion, the flow system described here creates complex, physiologically relevant flow profiles that provide deeper insight into the fluid dynamics of separated flow and pave the way for future studies interrogating the cellular response to complex flow regimes.


Asunto(s)
Barrera Hematoencefálica , Técnicas de Cultivo de Célula , Dispositivos Laboratorio en un Chip , Técnicas Analíticas Microfluídicas , Modelos Cardiovasculares , Uniones Estrechas/metabolismo , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/metabolismo , Humanos
8.
Neurobiol Dis ; 146: 105131, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33053430

RESUMEN

As researchers across the globe have focused their attention on understanding SARS-CoV-2, the picture that is emerging is that of a virus that has serious effects on the vasculature in multiple organ systems including the cerebral vasculature. Observed effects on the central nervous system include neurological symptoms (headache, nausea, dizziness), fatal microclot formation and in rare cases encephalitis. However, our understanding of how the virus causes these mild to severe neurological symptoms and how the cerebral vasculature is impacted remains unclear. Thus, the results presented in this report explored whether deleterious outcomes from the SARS-CoV-2 viral spike protein on primary human brain microvascular endothelial cells (hBMVECs) could be observed. The spike protein, which plays a key role in receptor recognition, is formed by the S1 subunit containing a receptor binding domain (RBD) and the S2 subunit. First, using postmortem brain tissue, we show that the angiotensin converting enzyme 2 or ACE2 (a known binding target for the SARS-CoV-2 spike protein), is ubiquitously expressed throughout various vessel calibers in the frontal cortex. Moreover, ACE2 expression was upregulated in cases of hypertension and dementia. ACE2 was also detectable in primary hBMVECs maintained under cell culture conditions. Analysis of cell viability revealed that neither the S1, S2 or a truncated form of the S1 containing only the RBD had minimal effects on hBMVEC viability within a 48 h exposure window. Introduction of spike proteins to invitro models of the blood-brain barrier (BBB) showed significant changes to barrier properties. Key to our findings is the demonstration that S1 promotes loss of barrier integrity in an advanced 3D microfluidic model of the human BBB, a platform that more closely resembles the physiological conditions at this CNS interface. Evidence provided suggests that the SARS-CoV-2 spike proteins trigger a pro-inflammatory response on brain endothelial cells that may contribute to an altered state of BBB function. Together, these results are the first to show the direct impact that the SARS-CoV-2 spike protein could have on brain endothelial cells; thereby offering a plausible explanation for the neurological consequences seen in COVID-19 patients.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Barrera Hematoencefálica/metabolismo , Permeabilidad Capilar/fisiología , Células Endoteliales/metabolismo , Inflamación/metabolismo , Metaloproteinasas de la Matriz/metabolismo , SARS-CoV-2/patogenicidad , Glicoproteína de la Espiga del Coronavirus/fisiología , Barrera Hematoencefálica/efectos de los fármacos , COVID-19 , Permeabilidad Capilar/efectos de los fármacos , Moléculas de Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Demencia/metabolismo , Impedancia Eléctrica , Células Endoteliales/efectos de los fármacos , Lóbulo Frontal/metabolismo , Humanos , Hipertensión/metabolismo , Técnicas In Vitro , Uniones Intercelulares/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Dispositivos Laboratorio en un Chip , Metaloproteinasas de la Matriz/efectos de los fármacos , Cultivo Primario de Células , Dominios Proteicos , Subunidades de Proteína/metabolismo , Subunidades de Proteína/farmacología , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Glicoproteína de la Espiga del Coronavirus/farmacología
9.
Exp Cell Res ; 376(1): 49-57, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30658092

RESUMEN

Astrocyte activation is crucial for wound contraction and glial scar formation following central nervous system injury, but the mechanism by which activation leads to astrocyte contractility and matrix reorganization in the central nervous system (CNS) is unknown. Current means to measure cell traction forces within three-dimensional scaffolds are limited to analyzing individual or small groups of cells, within extracellular matrices, whereas gap junctions and other cell-cell adhesions connect astrocytes to form a functional syncytium within the glial scar. Here, we measure the viscoelastic properties of cell-seeded hydrogels to yield insight into the collective contractility of astrocytes as they exert tension on the surrounding matrix and change its bulk mechanical properties. Our results indicate that incorporation of the CNS matrix component hyaluronan into a collagen hydrogel increases expression of the intermediate filament protein GFAP and results in a higher shear storage modulus of the cell/matrix composite, establishing the correlation between astrocyte activation and increased cell contractility. The effects of thrombin and blebbistatin, known mediators of actomyosin-mediated contraction, verify that cell-matrix tension dictates the hydrogel mechanical properties. Viability assays indicate that increased cell traction exacerbates cell death at the center of the scaffold, and message level analysis reveals that cells in the hyaluronan-containing matrix have a ~ 3-fold increase in HIF-1α gene expression. Overall, these findings suggest that astrocyte activation not only increases cell traction, but may also contribute to hypoxia near sites of central nervous system injury.


Asunto(s)
Sistema Nervioso Central/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Hidrogeles/farmacología , Neuroglía/efectos de los fármacos , Astrocitos/efectos de los fármacos , Técnicas de Cultivo de Célula , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/genética , Sistema Nervioso Central/lesiones , Cicatriz/tratamiento farmacológico , Cicatriz/patología , Colágeno/química , Colágeno/farmacología , Citosol/química , Humanos , Ácido Hialurónico/química , Ácido Hialurónico/farmacología , Hidrogeles/química , Neuroglía/patología , Oxígeno/metabolismo , Reología/métodos , Sustancias Viscoelásticas/química , Sustancias Viscoelásticas/farmacología , Cicatrización de Heridas/efectos de los fármacos
10.
Biophys J ; 116(7): 1340-1347, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30878203

RESUMEN

The extracellular matrix in vivo contains variable but often large amounts of glycosaminoglycans that influence cell and tissue function. Hyaluronan (HA) is an abundant glycosaminoglycan within the extracellular matrix of the myocardium during early development and in the aftermath of a myocardial infarction. Its flexible anionic structure has a strong influence on mechanical response and interstitial fluid flow within the matrix. Additionally, HA has a direct, biochemical effect on cells through an array of cell-surface receptors, including CD44, RHAMM/CD168, and other surface-exposed structures. Recent studies have shown that HA modulates the response of cardiomyocytes and other cell types to two-dimensional substrates of varying elastic moduli. This study investigates the force response to HA of cardiomyocytes and cardiac fibroblasts within three-dimensional matrices of variable composition and mechanical properties in vitro. HA significantly decreased the force exerted by the cell-matrix constructs in a tensiometer testing platform and within microfabricated tissue gauges. However, its effect was no different from that of alginate, an anionic polysaccharide with the same charge density but no specific transmembrane receptors. Therefore, these results establish that HA exerts a generic physical-chemical effect within three-dimensional hydrogels that must be accounted for when interrogating cell-matrix interactions.


Asunto(s)
Ácido Hialurónico/farmacología , Hidrogeles/química , Miocitos Cardíacos/efectos de los fármacos , Alginatos/química , Animales , Células Cultivadas , Matriz Extracelular/química , Fibrina/química , Ácido Hialurónico/química , Ratas , Ratas Sprague-Dawley , Electricidad Estática , Estrés Mecánico
11.
Microcirculation ; 26(7): e12576, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31140665

RESUMEN

OBJECTIVE: Low-intensity anti-vascular ultrasound therapy is an effective means of disrupting the blood supply in the tumor microenvironment. Its diminished effect on the surrounding vasculature is thought to be due to higher blood flow rates outside the tumor that decreases the interaction time between the endothelial lining and the microbubbles, which transduce acoustic energy to thermal heat. However, investigating the effect of circulation rate on the response to low-intensity ultrasound is complicated by the heterogeneity of the in vivo vascular microenvironment. Here, a 3D microfluidic model is used to directly interrogate the dynamics of ultrasound stimulation. METHODS: A 3D in vitro vessel consisting of LifeACT transfected endothelial cells facilitate real-time analysis of actin dynamics during ultrasound treatment. Using an integrated testing platform, both the flow rate of microbubbles within the vessel and the magnitude of insonation can be varied. RESULTS: Morphological measurements and dextran transport assays indicate that lower flow rates exacerbate the effect of low-intensity ultrasound on vessel integrity. Additionally, immunostaining for VE-cadherin and transmission electron microscopy provide further insight into structural changes in cell-cell junctions following insonation. CONCLUSIONS: Overall, these results reveal that blood flow rate is an important parameter to consider during the refinement of anti-vascular low-intensity ultrasound therapies.


Asunto(s)
Células Endoteliales/metabolismo , Microfluídica , Modelos Cardiovasculares , Neoplasias , Neovascularización Patológica , Microambiente Tumoral , Terapia por Ultrasonido , Antígenos CD/metabolismo , Cadherinas/metabolismo , Células Endoteliales/patología , Humanos , Neoplasias/irrigación sanguínea , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/terapia , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Neovascularización Patológica/terapia
12.
Small ; 15(50): e1903180, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31721440

RESUMEN

The migration of cells through constricting spaces or along fibrous tracks in tissues is important for many biological processes and depends on the mechanical properties of a cytoskeleton made up of three different filaments: F-actin, microtubules, and intermediate filaments. The signaling pathways and cytoskeletal structures that control cell motility on 2D are often very different from those that control motility in 3D. Previous studies have shown that intermediate filaments can promote actin-driven protrusions at the cell edge, but have little effect on overall motility of cells on flat surfaces. They are however important for cells to maintain resistance to repeated compressive stresses that are expected to occur in vivo. Using mouse embryonic fibroblasts derived from wild-type and vimentin-null mice, it is found that loss of vimentin increases motility in 3D microchannels even though on flat surfaces it has the opposite effect. Atomic force microscopy and traction force microscopy experiments reveal that vimentin enhances perinuclear cell stiffness while maintaining the same level of acto-myosin contractility in cells. A minimal model in which a perinuclear vimentin cage constricts along with the nucleus during motility through confining spaces, providing mechanical resistance against large strains that could damage the structural integrity of cells, is proposed.


Asunto(s)
Movimiento Celular , Vimentina/deficiencia , Animales , Fenómenos Biomecánicos , Capilares/efectos de los fármacos , Colágeno/farmacología , Citoesqueleto/metabolismo , Hidrogeles/farmacología , Ratones , Miosina Tipo II/metabolismo , Células 3T3 NIH , Vimentina/metabolismo
13.
Proc Natl Acad Sci U S A ; 111(22): 7968-73, 2014 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-24843171

RESUMEN

The density and architecture of capillary beds that form within a tissue depend on many factors, including local metabolic demand and blood flow. Here, using microfluidic control of local fluid mechanics, we show the existence of a previously unappreciated flow-induced shear stress threshold that triggers angiogenic sprouting. Both intraluminal shear stress over the endothelium and transmural flow through the endothelium above 10 dyn/cm(2) triggered endothelial cells to sprout and invade into the underlying matrix, and this threshold is not impacted by the maturation of cell-cell junctions or pressure gradient across the monolayer. Antagonizing VE-cadherin widened cell-cell junctions and reduced the applied shear stress for a given transmural flow rate, but did not affect the shear threshold for sprouting. Furthermore, both transmural and luminal flow induced expression of matrix metalloproteinase 1, and this up-regulation was required for the flow-induced sprouting. Once sprouting was initiated, continuous flow was needed to both sustain sprouting and prevent retraction. To explore the potential ramifications of a shear threshold on the spatial patterning of new sprouts, we used finite-element modeling to predict fluid shear in a variety of geometric settings and then experimentally demonstrated that transmural flow guided preferential sprouting toward paths of draining interstitial fluid flow as might occur to connect capillary beds to venules or lymphatics. In addition, we show that luminal shear increases in local narrowings of vessels to trigger sprouting, perhaps ultimately to normalize shear stress across the vasculature. Together, these studies highlight the role of shear stress in controlling angiogenic sprouting and offer a potential homeostatic mechanism for regulating vascular density.


Asunto(s)
Células Endoteliales/fisiología , Metaloproteinasa 1 de la Matriz/fisiología , Microfluídica/instrumentación , Microfluídica/métodos , Modelos Biológicos , Neovascularización Fisiológica/fisiología , Adenoviridae/genética , Capilares/citología , Capilares/fisiología , Movimiento Celular/fisiología , Células Endoteliales/citología , Células Endoteliales/ultraestructura , Análisis de Elementos Finitos , Silenciador del Gen , Células Endoteliales de la Vena Umbilical Humana , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Metaloproteinasa 1 de la Matriz/genética , Mecanotransducción Celular/fisiología , Microscopía Electrónica de Transmisión , Estrés Mecánico
14.
Soft Matter ; 12(22): 5050-60, 2016 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-27174568

RESUMEN

We present theoretical and experimental studies of the elastic response of fibrous networks subjected to uniaxial strain. Uniaxial compression or extension is applied to extracellular networks of fibrin and collagen using a shear rheometer with free water in/outflow. Both uniaxial stress and the network shear modulus are measured. Prior work [van Oosten, et al., Sci. Rep., 2015, 6, 19270] has shown softening/stiffening of these networks under compression/extension, together with a nonlinear response to shear, but the origin of such behaviour remains poorly understood. Here, we study how uniaxial strain influences the nonlinear mechanics of fibrous networks. Using a computational network model with bendable and stretchable fibres, we show that the softening/stiffening behaviour can be understood for fixed lateral boundaries in 2D and 3D networks with comparable average connectivities to the experimental extracellular networks. Moreover, we show that the onset of stiffening depends strongly on the imposed uniaxial strain. Our study highlights the importance of both uniaxial strain and boundary conditions in determining the mechanical response of hydrogels.

15.
Proc Natl Acad Sci U S A ; 110(17): 6712-7, 2013 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-23569284

RESUMEN

Angiogenesis is a complex morphogenetic process whereby endothelial cells from existing vessels invade as multicellular sprouts to form new vessels. Here, we have engineered a unique organotypic model of angiogenic sprouting and neovessel formation that originates from preformed artificial vessels fully encapsulated within a 3D extracellular matrix. Using this model, we screened the effects of angiogenic factors and identified two distinct cocktails that promoted robust multicellular endothelial sprouting. The angiogenic sprouts in our system exhibited hallmark structural features of in vivo angiogenesis, including directed invasion of leading cells that developed filopodia-like protrusions characteristic of tip cells, following stalk cells exhibiting apical-basal polarity, and lumens and branches connecting back to the parent vessels. Ultimately, sprouts bridged between preformed channels and formed perfusable neovessels. Using this model, we investigated the effects of angiogenic inhibitors on sprouting morphogenesis. Interestingly, the ability of VEGF receptor 2 inhibition to antagonize filopodia formation in tip cells was context-dependent, suggesting a mechanism by which vessels might be able to toggle between VEGF-dependent and VEGF-independent modes of angiogenesis. Like VEGF, sphingosine-1-phosphate also seemed to exert its proangiogenic effects by stimulating directional filopodial extension, whereas matrix metalloproteinase inhibitors prevented sprout extension but had no impact on filopodial formation. Together, these results demonstrate an in vitro 3D biomimetic model that reconstitutes the morphogenetic steps of angiogenic sprouting and highlight the potential utility of the model to elucidate the molecular mechanisms that coordinate the complex series of events involved in neovascularization.


Asunto(s)
Biomimética/métodos , Microfluídica/métodos , Modelos Biológicos , Morfogénesis/fisiología , Neovascularización Fisiológica/fisiología , Polaridad Celular/fisiología , Dimetilpolisiloxanos , Clorhidrato de Fingolimod , Técnica del Anticuerpo Fluorescente , Células Endoteliales de la Vena Umbilical Humana , Humanos , Indoles/farmacología , Lisofosfolípidos/metabolismo , Morfogénesis/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Glicoles de Propileno/farmacología , Seudópodos/fisiología , Pirroles/farmacología , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Esfingosina/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores
16.
Cytotherapy ; 16(7): 906-14, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24713331

RESUMEN

BACKGROUND AIMS: Myocardial infarction results in the formation of scar tissue populated by myofibroblasts, a phenotype characterized by increased contractility and matrix deposition. Mesenchymal stromal cells (MSC) delivered to the myocardium can attenuate scar growth and restore cardiac function, though the mechanism is unclear. METHODS: This study describes a simple yet robust three-dimensional (3D) in vitro co-culture model to examine the paracrine effects of implanted MSC on resident myofibroblasts in a controlled biochemical and mechanical environment. The fibrosis model consisted of fibroblasts embedded in a 3D collagen gel cultured under defined oxygen tensions and exposed to either cyclic strain or interstitial fluid flow. MSC were injected into this model, and the effect on fibroblast phenotype was evaluated 48 h after cell injection. RESULTS: Analysis of gene and protein expression of the fibroblasts indicated that injection of MSC attenuated the myofibroblast transition in response to reduced oxygen and mechanical stress. Assessment of vascular endothelial growth factor and insulin-like growth factor-1 levels demonstrated that their release by fibroblasts was markedly upregulated in hypoxic conditions but attenuated by strain or fluid flow. In fibroblast-MSC co-cultures, vascular endothelial growth factor levels were increased by hypoxia but not affected by mechanical stimuli, whereas insulin-like growth factor-1 levels were generally low and not affected by experimental conditions. CONCLUSIONS: This study demonstrates how a 3D in vitro model of the cardiac scar can be used to examine paracrine effects of MSC on the phenotype of resident fibroblasts and therefore illuminates the role of injected progenitor cells on the progression of cardiac fibrosis.


Asunto(s)
Diferenciación Celular , Células Madre Mesenquimatosas/citología , Infarto del Miocardio/metabolismo , Estrés Mecánico , Animales , Hipoxia de la Célula/genética , Cicatriz/patología , Técnicas de Cocultivo , Fibrosis , Humanos , Técnicas In Vitro , Células Madre Mesenquimatosas/metabolismo , Infarto del Miocardio/patología , Miofibroblastos/patología , Oxígeno/metabolismo , Comunicación Paracrina/genética , Ratas , Somatomedinas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
17.
ACS Appl Mater Interfaces ; 16(12): 14457-14466, 2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38488736

RESUMEN

Most in vitro models use culture medium to apply fluid shear stress to endothelial cells, which does not capture the interaction between blood and endothelial cells. Here, we describe a new system to characterize whole blood flow through a 3D-printed, endothelialized vascular topology that induces flow separation at a bifurcation. Drag-reducing polymers, which have been previously studied as a potential therapy to reduce the pressure drop across the vascular bed, are evaluated for their effect on mitigating the disturbed flow. Polymer concentrations of 1000 ppm prevented recirculation and disturbed flow at the wall. Proteomic analysis of plasma collected from whole blood recirculated through the vascularized channel with and without drag-reducing polymers provides insight into the effects of flow regimes on levels of proteins indicative of the endothelial-blood interaction. The results indicate that blood flow alters proteins associated with coagulation, inflammation, and other processes. Overall, these proof-of-concept experiments demonstrate the importance of using whole blood flow to study the endothelial response to perfusion.


Asunto(s)
Células Endoteliales , Polímeros , Polímeros/farmacología , Proteómica , Hemodinámica/fisiología , Impresión Tridimensional , Estrés Mecánico
18.
SLAS Technol ; 29(2): 100121, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38340892

RESUMEN

A major aim in the field of synthetic biology is developing tools capable of responding to user-defined inputs by activating therapeutically relevant cellular functions. Gene transcription and regulation in response to external stimuli are some of the most powerful and versatile of these cellular functions being explored. Motivated by the success of chimeric antigen receptor (CAR) T-cell therapies, transmembrane receptor-based platforms have been embraced for their ability to sense extracellular ligands and to subsequently activate intracellular signal transduction. The integration of transmembrane receptors with transcriptional activation platforms has not yet achieved its full potential. Transient expression of plasmid DNA is often used to explore gene regulation platforms in vitro. However, applications capable of targeting therapeutically relevant endogenous or stably integrated genes are more clinically relevant. Gene regulation may allow for engineered cells to traffic into tissues of interest and secrete functional proteins into the extracellular space or to differentiate into functional cells. Transmembrane receptors that regulate transcription have the potential to revolutionize cell therapies in a myriad of applications, including cancer treatment and regenerative medicine. In this review, we will examine current engineering approaches to control transcription in mammalian cells with an emphasis on systems that can be selectively activated in response to extracellular signals. We will also speculate on the potential therapeutic applications of these technologies and examine promising approaches to expand their capabilities and tighten the control of gene regulation in cellular therapies.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Regulación de la Expresión Génica , Animales , Transducción de Señal , Ligandos , Mamíferos
19.
Biophys J ; 105(9): 2188-98, 2013 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-24209865

RESUMEN

In the creation of engineered tissue constructs, the successful transport of nutrients and oxygen to the contained cells is a significant challenge. In highly porous scaffolds subject to cyclic strain, the mechanical deformations can induce substantial fluid pressure gradients, which affect the transport of solutes. In this article, we describe a poroelastic model to predict the solid and fluid mechanics of a highly porous hydrogel subject to cyclic strain. The model was validated by matching the predicted penetration of a bead into the hydrogel from the model with experimental observations and provides insight into nutrient transport. Additionally, the model provides estimates of the wall-shear stresses experienced by the cells embedded within the scaffold. These results provide insight into the mechanics of and convective nutrient transport within a cyclically strained hydrogel, which could lead to the improved design of engineered tissues.


Asunto(s)
Colágeno/química , Colágeno/metabolismo , Elasticidad , Hidrogeles , Modelos Biológicos , Estrés Mecánico , Transporte Biológico , Porosidad
20.
Nat Mater ; 11(9): 768-74, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22751181

RESUMEN

In the absence of perfusable vascular networks, three-dimensional (3D) engineered tissues densely populated with cells quickly develop a necrotic core. Yet the lack of a general approach to rapidly construct such networks remains a major challenge for 3D tissue culture. Here, we printed rigid 3D filament networks of carbohydrate glass, and used them as a cytocompatible sacrificial template in engineered tissues containing living cells to generate cylindrical networks that could be lined with endothelial cells and perfused with blood under high-pressure pulsatile flow. Because this simple vascular casting approach allows independent control of network geometry, endothelialization and extravascular tissue, it is compatible with a wide variety of cell types, synthetic and natural extracellular matrices, and crosslinking strategies. We also demonstrated that the perfused vascular channels sustained the metabolic function of primary rat hepatocytes in engineered tissue constructs that otherwise exhibited suppressed function in their core.


Asunto(s)
Vasos Sanguíneos/citología , Perfusión , Impresión/métodos , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Animales , Materiales Biocompatibles/química , Materiales Biocompatibles/metabolismo , Materiales Biocompatibles/farmacología , Carbohidratos/química , Vidrio/química , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Ratas , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA