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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(41): e2308941120, 2023 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-37782785

RESUMEN

Impaired lymphatic drainage and lymphedema are major morbidities whose mechanisms have remained obscure. To study lymphatic drainage and its impairment, we engineered a microfluidic culture model of lymphatic vessels draining interstitial fluid. This lymphatic drainage-on-chip revealed that inflammatory cytokines that are known to disrupt blood vessel junctions instead tightened lymphatic cell-cell junctions and impeded lymphatic drainage. This opposing response was further demonstrated when inhibition of rho-associated protein kinase (ROCK) was found to normalize fluid drainage under cytokine challenge by simultaneously loosening lymphatic junctions and tightening blood vessel junctions. Studies also revealed a previously undescribed shift in ROCK isoforms in lymphatic endothelial cells, wherein a ROCK2/junctional adhesion molecule-A (JAM-A) complex emerges that is responsible for the cytokine-induced lymphatic junction zippering. To validate these in vitro findings, we further demonstrated in a genetic mouse model that lymphatic-specific knockout of ROCK2 reversed lymphedema in vivo. These studies provide a unique platform to generate interstitial fluid pressure and measure the drainage of interstitial fluid into lymphatics and reveal a previously unappreciated ROCK2-mediated mechanism in regulating lymphatic drainage.


Asunto(s)
Molécula A de Adhesión de Unión , Vasos Linfáticos , Linfedema , Quinasas Asociadas a rho , Animales , Ratones , Biomimética , Citocinas/metabolismo , Células Endoteliales/metabolismo , Uniones Intercelulares , Molécula A de Adhesión de Unión/metabolismo , Vasos Linfáticos/metabolismo , Linfedema/genética , Linfedema/metabolismo , Quinasas Asociadas a rho/metabolismo
2.
Proc Natl Acad Sci U S A ; 119(28): e2115867119, 2022 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-35763565

RESUMEN

Liver regeneration is a well-orchestrated process that is typically studied in animal models. Although previous animal studies have offered many insights into liver regeneration, human biology is less well understood. To this end, we developed a three-dimensional (3D) platform called structurally vascularized hepatic ensembles for analyzing regeneration (SHEAR) to model multiple aspects of human liver regeneration. SHEAR enables control over hemodynamic alterations to mimic those that occur during liver injury and regeneration and supports the administration of biochemical inputs such as cytokines and paracrine interactions with endothelial cells. We found that exposing the endothelium-lined channel to fluid flow led to increased secretion of regeneration-associated factors. Stimulation with relevant cytokines not only amplified the secretory response, but also induced cell-cycle entry of primary human hepatocytes (PHHs) embedded within the device. Further, we identified endothelial-derived mediators that are sufficient to initiate proliferation of PHHs in this context. Collectively, the data presented here underscore the importance of multicellular models that can recapitulate high-level tissue functions and demonstrate that the SHEAR device can be used to discover and validate conditions that promote human liver regeneration.


Asunto(s)
Células Endoteliales , Hepatocitos , Regeneración Hepática , Hígado , Técnicas de Cultivo Tridimensional de Células , Citocinas , Humanos , Hígado/irrigación sanguínea , Regeneración Hepática/fisiología
3.
Small ; 20(24): e2307901, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38185718

RESUMEN

Cardiovascular disease is the cause of death in ≈50% of hemodialysis patients. Accumulation of uremic solutes in systemic circulation is thought to be a key driver of the endothelial dysfunction that underlies elevated cardiovascular events. A challenge in understanding the mechanisms relating chronic kidney disease to cardiovascular disease is the lack of in vitro models that allow screening of the effects of the uremic environment on the endothelium. Here, a method is described for microfabrication of human blood vessels from donor cells and perfused with donor serum. The resulting donor-derived microvessels are used to quantify vascular permeability, a hallmark of endothelial dysfunction, in response to serum spiked with pathophysiological levels of indoxyl sulfate, and in response to serum from patients with chronic kidney disease and from uremic pigs. The uremic environment has pronounced effects on microvascular integrity as demonstrated by irregular cell-cell junctions and increased permeability in comparison to cell culture media and healthy serum. Moreover, the engineered microvessels demonstrate an increase in sensitivity compared to traditional 2D assays. Thus, the devices and the methods presented here have the potential to be utilized to risk stratify and to direct personalized treatments for patients with chronic kidney disease.


Asunto(s)
Enfermedades Cardiovasculares , Microvasos , Humanos , Microvasos/patología , Animales , Porcinos , Insuficiencia Renal/terapia , Medición de Riesgo , Donantes de Tejidos , Ingeniería de Tejidos/métodos , Indicán/sangre
4.
Nature ; 552(7684): 258-262, 2017 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-29160307

RESUMEN

The vascular barrier that separates blood from tissues is actively regulated by the endothelium and is essential for transport, inflammation, and haemostasis. Haemodynamic shear stress plays a critical role in maintaining endothelial barrier function, but how this occurs remains unknown. Here we use an engineered organotypic model of perfused microvessels to show that activation of the transmembrane receptor NOTCH1 directly regulates vascular barrier function through a non-canonical, transcription-independent signalling mechanism that drives assembly of adherens junctions, and confirm these findings in mouse models. Shear stress triggers DLL4-dependent proteolytic activation of NOTCH1 to expose the transmembrane domain of NOTCH1. This domain mediates establishment of the endothelial barrier; expression of the transmembrane domain of NOTCH1 is sufficient to rescue defects in barrier function induced by knockout of NOTCH1. The transmembrane domain restores barrier function by catalysing the formation of a receptor complex in the plasma membrane consisting of vascular endothelial cadherin, the transmembrane protein tyrosine phosphatase LAR, and the RAC1 guanidine-exchange factor TRIO. This complex activates RAC1 to drive assembly of adherens junctions and establish barrier function. Canonical transcriptional signalling via Notch is highly conserved in metazoans and is required for many processes in vascular development, including arterial-venous differentiation, angiogenesis and remodelling. We establish the existence of a non-canonical cortical NOTCH1 signalling pathway that regulates vascular barrier function, and thus provide a mechanism by which a single receptor might link transcriptional programs with adhesive and cytoskeletal remodelling.


Asunto(s)
Uniones Adherentes/metabolismo , Endotelio Vascular/metabolismo , Complejos Multiproteicos/metabolismo , Receptor Notch1/metabolismo , Uniones Adherentes/enzimología , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Línea Celular , Endotelio Vascular/enzimología , Femenino , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Ratones , Complejos Multiproteicos/química , Fosfoproteínas/metabolismo , Dominios Proteicos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Receptor Notch1/química , Transducción de Señal , Proteínas de Unión al GTP rac/metabolismo
5.
Development ; 145(9)2018 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-29712669

RESUMEN

The architecture of individual cells and cell collectives enables functional specification, a prominent example being the formation of epithelial tubes that transport fluid or gas in many organs. The intrahepatic bile ducts (IHBDs) form a tubular network within the liver parenchyma that transports bile to the intestine. Aberrant biliary 'neoductulogenesis' is also a feature of several liver pathologies including tumorigenesis. However, the mechanism of biliary tube morphogenesis in development or disease is not known. Elimination of the neurofibromatosis type 2 protein (NF2; also known as merlin or neurofibromin 2) causes hepatomegaly due to massive biliary neoductulogenesis in the mouse liver. We show that this phenotype reflects unlimited biliary morphogenesis rather than proliferative expansion. Our studies suggest that NF2 normally limits biliary morphogenesis by coordinating lumen expansion and cell architecture. This work provides fundamental insight into how biliary fate and tubulogenesis are coordinated during development and will guide analyses of disease-associated and experimentally induced biliary pathologies.


Asunto(s)
Conductos Biliares Intrahepáticos/embriología , Proliferación Celular/fisiología , Neurofibromina 2/metabolismo , Organogénesis/fisiología , Animales , Conductos Biliares Intrahepáticos/patología , Eliminación de Gen , Hepatomegalia/embriología , Hepatomegalia/genética , Hepatomegalia/patología , Ratones , Ratones Noqueados , Neurofibromina 2/genética
6.
Hepatology ; 71(4): 1350-1363, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31465556

RESUMEN

BACKGROUND AND AIMS: Chronic cholestatic liver diseases, such as primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC), are frequently associated with damage to the barrier function of the biliary epithelium. Here, we report on a bile duct-on-a-chip that phenocopies not only the tubular architecture of the bile duct in three dimensions, but also its barrier functions. APPROACH AND RESULTS: We showed that mouse cholangiocytes in the channel of the device became polarized and formed mature tight junctions, that the permeability of the cholangiocyte monolayer was comparable to ex vivo measurements, and that cholangiocytes in the device were mechanosensitive (as demonstrated by changes in calcium flux under applied luminal flow). Permeability decreased significantly when cells formed a compact monolayer with cell densities comparable to those observed in vivo. This device enabled independent access to the apical and basolateral surfaces of the cholangiocyte channel, allowing proof-of-concept toxicity studies with the biliary toxin, biliatresone, and the bile acid, glycochenodeoxycholic acid. The cholangiocyte basolateral side was more vulnerable than the apical side to treatment with either agent, suggesting a protective adaptation of the apical surface that is normally exposed to bile. Further studies revealed a protective role of the cholangiocyte apical glycocalyx, wherein disruption of the glycocalyx with neuraminidase increased the permeability of the cholangiocyte monolayer after treatment with glycochenodeoxycholic acid. CONCLUSIONS: This bile duct-on-a-chip captured essential features of a simplified bile duct in structure and organ-level functions and represents an in vitro platform to study the pathophysiology of the bile duct using cholangiocytes from a variety of sources.


Asunto(s)
Conductos Biliares/fisiopatología , Dispositivos Laboratorio en un Chip , Animales , Línea Celular , Células Epiteliales , Ratones , Ratones Endogámicos BALB C , Modelos Animales
7.
J Phys D Appl Phys ; 53(22)2020 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-33840837

RESUMEN

Mechanical forces regulate a diverse set of biological processes at cellular, tissue, and organismal length scales. Investigating the cellular and molecular mechanisms that underlie the conversion of mechanical forces to biological responses is challenged by limitations of traditional animal models and in vitro cell culture, including poor control over applied force and highly artificial cell culture environments. Recent advances in fabrication methods and material processing have enabled the development of microfluidic platforms that provide precise control over the mechanical microenvironment of cultured cells. These devices and systems have proven to be powerful for uncovering and defining mechanisms of mechanotransduction. In this review, we first give an overview of the main mechanotransduction pathways that function at sites of cell adhesion, many of which have been investigated with microfluidics. We then discuss how distinct microfluidic fabrication methods can be harnessed to gain biological insight, with description of both monolithic and replica molding approaches. Finally, we present examples of how microfluidics can be used to apply both solid forces (substrate mechanics, strain, and compression) and fluid forces (luminal, interstitial) to cells. Throughout the review, we emphasize the advantages and disadvantages of different fabrication methods and applications of force in order to provide perspective to investigators looking to apply forces to cells in their own research.

8.
Proc Natl Acad Sci U S A ; 114(33): 8758-8763, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28765370

RESUMEN

The integrity of the endothelial barrier between circulating blood and tissue is important for blood vessel function and, ultimately, for organ homeostasis. Here, we developed a vessel-on-a-chip with perfused endothelialized channels lined with human bone marrow stromal cells, which adopt a mural cell-like phenotype that recapitulates barrier function of the vasculature. In this model, barrier function is compromised upon exposure to inflammatory factors such as LPS, thrombin, and TNFα, as has been observed in vivo. Interestingly, we observed a rapid physical withdrawal of mural cells from the endothelium that was accompanied by an inhibition of endogenous Rac1 activity and increase in RhoA activity in the mural cells themselves upon inflammation. Using a system to chemically induce activity in exogenously expressed Rac1 or RhoA within minutes of stimulation, we demonstrated RhoA activation induced loss of mural cell coverage on the endothelium and reduced endothelial barrier function, and this effect was abrogated when Rac1 was simultaneously activated. We further showed that N-cadherin expression in mural cells plays a key role in barrier function, as CRISPR-mediated knockout of N-cadherin in the mural cells led to loss of barrier function, and overexpression of N-cadherin in CHO cells promoted barrier function. In summary, this bicellular model demonstrates the continuous and rapid modulation of adhesive interactions between endothelial and mural cells and its impact on vascular barrier function and highlights an in vitro platform to study the biology of perivascular-endothelial interactions.


Asunto(s)
Cadherinas/metabolismo , Endotelio Vascular/metabolismo , Endotelio/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Biomimética/métodos , Células CHO , Cricetulus , Humanos , Inflamación/metabolismo , Trombina/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
9.
Nat Methods ; 13(5): 415-23, 2016 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-27123817

RESUMEN

Forces generated by cells are critical regulators of cell adhesion, signaling, and function, and they are also essential drivers in the morphogenetic events of development. Over the past 20 years, several methods have been developed to measure these forces. However, despite recent substantial interest in understanding the contribution of these forces in biology, implementation and adoption of the developed methods by the broader biological community remain challenging because of the inherently multidisciplinary expertise required to conduct and interpret the measurements. In this review, we introduce the established methods and highlight the technical challenges associated with implementing each technique in a biological laboratory.


Asunto(s)
Fenómenos Biomecánicos , Fenómenos Biofísicos , Adhesión Celular/fisiología , Matriz Extracelular/fisiología , Modelos Biológicos , Movimiento Celular/fisiología , Células Cultivadas , Módulo de Elasticidad/fisiología , Microscopía de Fuerza Atómica
10.
Nat Methods ; 12(12): 1132-4, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26436482

RESUMEN

Current measurements of the biomechanical properties of cells require physical contact with cells or lack subcellular resolution. Here we developed a label-free microscopy technique based on Brillouin light scattering that is capable of measuring an intracellular longitudinal modulus with optical resolution. The 3D Brillouin maps we obtained of cells in 2D and 3D microenvironments revealed mechanical changes due to cytoskeletal modulation and cell-volume regulation.


Asunto(s)
Citoesqueleto/química , Matriz Extracelular/química , Microscopía Confocal/métodos , Animales , Fenómenos Biomecánicos , Tamaño de la Célula , Citoesqueleto/ultraestructura , Módulo de Elasticidad , Matriz Extracelular/ultraestructura , Imagenología Tridimensional , Ratones , Microscopía Confocal/instrumentación , Células 3T3 NIH , Presión Osmótica
11.
J Am Soc Nephrol ; 28(6): 1741-1752, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28062569

RESUMEN

Ischemia-reperfusion injury (IRI) is a leading cause of AKI. This common clinical complication lacks effective therapies and can lead to the development of CKD. The αvß5 integrin may have an important role in acute injury, including septic shock and acute lung injury. To examine its function in AKI, we utilized a specific function-blocking antibody to inhibit αvß5 in a rat model of renal IRI. Pretreatment with this anti-αvß5 antibody significantly reduced serum creatinine levels, diminished renal damage detected by histopathologic evaluation, and decreased levels of injury biomarkers. Notably, therapeutic treatment with the αvß5 antibody 8 hours after IRI also provided protection from injury. Global gene expression profiling of post-ischemic kidneys showed that αvß5 inhibition affected established injury markers and induced pathway alterations previously shown to be protective. Intravital imaging of post-ischemic kidneys revealed reduced vascular leak with αvß5 antibody treatment. Immunostaining for αvß5 in the kidney detected evident expression in perivascular cells, with negligible expression in the endothelium. Studies in a three-dimensional microfluidics system identified a pericyte-dependent role for αvß5 in modulating vascular leak. Additional studies showed αvß5 functions in the adhesion and migration of kidney pericytes in vitro Initial studies monitoring renal blood flow after IRI did not find significant effects with αvß5 inhibition; however, future studies should explore the contribution of vasomotor effects. These studies identify a role for αvß5 in modulating injury-induced renal vascular leak, possibly through effects on pericyte adhesion and migration, and reveal αvß5 inhibition as a promising therapeutic strategy for AKI.


Asunto(s)
Permeabilidad Capilar/efectos de los fármacos , Riñón/irrigación sanguínea , Receptores de Vitronectina/antagonistas & inhibidores , Daño por Reperfusión/prevención & control , Animales , Masculino , Ratas , Ratas Sprague-Dawley
12.
Hepatology ; 73(2): 872-873, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33128466
13.
Proc Natl Acad Sci U S A ; 111(7): 2447-52, 2014 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-24550267

RESUMEN

Solid tumors are characterized by high interstitial fluid pressure, which drives fluid efflux from the tumor core. Tumor-associated interstitial flow (IF) at a rate of ∼3 µm/s has been shown to induce cell migration in the upstream direction (rheotaxis). However, the molecular biophysical mechanism that underlies upstream cell polarization and rheotaxis remains unclear. We developed a microfluidic platform to investigate the effects of IF fluid stresses imparted on cells embedded within a collagen type I hydrogel, and we demonstrate that IF stresses result in a transcellular gradient in ß1-integrin activation with vinculin, focal adhesion kinase (FAK), FAK(PY397), F actin, and paxillin-dependent protrusion formation localizing to the upstream side of the cell, where matrix adhesions are under maximum tension. This previously unknown mechanism is the result of a force balance between fluid drag on the cell and matrix adhesion tension and is therefore a fundamental, but previously unknown, stimulus for directing cell movement within porous extracellular matrix.


Asunto(s)
Movimiento Celular/fisiología , Líquido Extracelular/metabolismo , Matriz Extracelular/metabolismo , Hidrodinámica , Mecanotransducción Celular/fisiología , Modelos Biológicos , Línea Celular Tumoral , Adhesiones Focales/fisiología , Proteínas Fluorescentes Verdes , Humanos , Integrina beta1/metabolismo , ARN Interferente Pequeño/genética , Transfección , Vinculina
14.
Cell Mol Life Sci ; 70(8): 1335-56, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22926411

RESUMEN

Tumor cell migration is essential for invasion and dissemination from primary solid tumors and for the establishment of lethal secondary metastases at distant organs. In vivo and in vitro models enabled identification of different factors in the tumor microenvironment that regulate tumor progression and metastasis. However, the mechanisms by which tumor cells integrate these chemical and mechanical signals from multiple sources to navigate the complex microenvironment remain poorly understood. In this review, we discuss the factors that influence tumor cell migration with a focus on the migration of transformed carcinoma cells. We provide an overview of the experimental and computational methods that allow the investigation of tumor cell migration, and we highlight the benefits and shortcomings of the various assays. We emphasize that the chemical and mechanical stimulus paradigms are not independent and that crosstalk between them motivates the development of new assays capable of applying multiple, simultaneous stimuli and imaging the cellular migratory response in real-time. These next-generation assays will more closely mimic the in vivo microenvironment to provide new insights into tumor progression, inform techniques to control tumor cell migration, and render cancer more treatable.


Asunto(s)
Movimiento Celular , Invasividad Neoplásica/patología , Neoplasias/patología , Microambiente Tumoral , Animales , Humanos , Mecanotransducción Celular , Neoplasias/metabolismo
15.
Proc Natl Acad Sci U S A ; 108(27): 11115-20, 2011 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-21690404

RESUMEN

Interstitial flow is the convective transport of fluid through tissue extracellular matrix. This creeping fluid flow has been shown to affect the morphology and migration of cells such as fibroblasts, cancer cells, endothelial cells, and mesenchymal stem cells. A microfluidic cell culture system was designed to apply stable pressure gradients and fluid flow and allow direct visualization of transient responses of cells seeded in a 3D collagen type I scaffold. We used this system to examine the effects of interstitial flow on cancer cell morphology and migration and to extend previous studies showing that interstitial flow increases the metastatic potential of MDA-MB-435S melanoma cells [Shields J, et al. (2007) Cancer Cell 11:526-538]. Using a breast carcinoma line (MDA-MB-231) we also observed cell migration along streamlines in the presence of flow; however, we further demonstrated that the strength of the flow as well as the cell density determined directional bias of migration along the streamline. In particular, we found that cells either at high seeding density or with the CCR-7 receptor inhibited migration against, rather than with the flow. We provide further evidence that CCR7-dependent autologous chemotaxis is the mechanism that leads to migration with the flow, but also demonstrate a competing CCR7-independent mechanism that causes migration against the flow. Data from experiments investigating the effects of cell concentration, interstitial flow rate, receptor activity, and focal adhesion kinase phosphorylation support our hypothesis that the competing stimulus is integrin mediated. This mechanism may play an important role in development of metastatic disease.


Asunto(s)
Neoplasias de la Mama/patología , Neoplasias de la Mama/fisiopatología , Movimiento Celular/fisiología , Ingeniería Biomédica , Neoplasias de la Mama/secundario , Recuento de Células , Línea Celular Tumoral , Quimiotaxis/fisiología , Líquido Extracelular/fisiología , Femenino , Análisis de Elementos Finitos , Quinasa 1 de Adhesión Focal/fisiología , Humanos , Técnicas Analíticas Microfluídicas , Modelos Biológicos , Metástasis de la Neoplasia/patología , Metástasis de la Neoplasia/fisiopatología , Receptores CCR7/antagonistas & inhibidores , Receptores CCR7/fisiología , Transducción de Señal
16.
Expert Opin Drug Discov ; 19(3): 339-351, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38117223

RESUMEN

INTRODUCTION: Vascular diseases impart a tremendous burden on healthcare systems in the United States and across the world. Efforts to improve therapeutic interventions are hindered by limitations of current experimental models. The integration of patient-derived cells with organ-on-chip (OoC) technology is a promising avenue for preclinical drug screening that improves upon traditional cell culture and animal models. AREAS COVERED: The authors review induced pluripotent stem cells (iPSC) and blood outgrowth endothelial cells (BOEC) as two sources for patient-derived endothelial cells (EC). They summarize several studies that leverage patient-derived EC and OoC for precision disease modeling of the vasculature, with a focus on applications for drug discovery. They also highlight the utility of patient-derived EC in other translational endeavors, including ex vivo organogenesis and multi-organ-chip integration. EXPERT OPINION: Precision disease modeling continues to mature in the academic space, but end-use by pharmaceutical companies is currently limited. To fully realize their transformative potential, OoC systems must balance their complexity with their ability to integrate with the highly standardized and high-throughput experimentation required for drug discovery and development.


Asunto(s)
Células Endoteliales , Células Madre Pluripotentes Inducidas , Animales , Humanos , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Dispositivos Laboratorio en un Chip
17.
ACS Appl Mater Interfaces ; 16(36): 47075-47088, 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39196896

RESUMEN

The tumor-vascular interface is a critical component of the tumor microenvironment that regulates all of the dynamic interactions between a growing tumor and the endothelial lining of the surrounding vasculature. In this paper, we report the design and development of a custom-engineered tumor-vascular interface system for investigating the early stage tumor-mediated pro-angiogenic dysfunctional behavior of the endothelium. Using representative endothelial cells and triple negative breast cancer cell lines, we established a biomimetic interface between a three-dimensional tumor tissue across a mature, functional endothelial barrier using a magnetically hybrid-integrated tumor-vascular interface system, wherein vasculature-like features containing a monolayer of endothelial cell culture on porous microfluidic channel surfaces were magnetically attached to tumor spheroids generated on a composite polymer-hydrogel microwell plate and embedded in a collagen matrix. Tumor-mediated endothelial microdynamics were characterized by their hallmark behavior such as loss of endothelial adherens junctions, increased cell density, proliferation, and changes in cell spreading and corroborated with endothelial YAP/TAZ nuclear translocation. We further confirm the feasibility of drug-mediated reversal of this pro-angiogenic endothelial organization through two different signaling mechanisms, namely, inhibition of the vascular endothelial growth factor pathway and the Notch signaling pathway, thereby demonstrating the utility of the tumor-vascular interface platform for rapid, early stage prediction of antiangiogenic drug efficacy. Overall, our work emphasizes the importance of our strategic engineering approach for identifying some unique, physiologically relevant aspects of the tumor-vascular interface, which are otherwise difficult to implement using standard in vitro approaches.


Asunto(s)
Dispositivos Laboratorio en un Chip , Humanos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Línea Celular Tumoral , Células Endoteliales de la Vena Umbilical Humana , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Microambiente Tumoral/efectos de los fármacos
18.
J Biomech ; 162: 111887, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38128469

RESUMEN

The high water content of articular cartilage allows this biphasic tissue to withstand large compressive loads through fluid pressurization. The system presented here, termed the "MagnaSquish", provides new capabilities for quantifying the effect of rehydration on cartilage behavior during cyclic loading. An imbalanced rate of fluid exudation during load and fluid re-entry during recovery can lead to the accumulation of strain during successive loading cycles - a phenomenon known as ratcheting. Typical experimental systems for cartilage biomechanics use continuous contact between the platen and sample, which may affect tissue rehydration by compressing the top layer of cartilage and slowing fluid re-entry. To address this limitation, we developed a magnetically actuated device that provides full lift-off of the platen in between loading cycles. We investigated strain accumulation in cadaveric human osteochondral plugs during 750 loading cycles, with two dimensional profiles of the cartilage captured at 30 frames per second throughout loading and 10 min of additional free swelling recovery. Axial and lateral strain measurements were extracted from the tissue profiles using a UNet-based deep learning algorithm to circumvent manual tracing. We observed increased axial strain accumulation with shorter inter-cycle recovery, with static loading serving as the extreme case of zero recovery. The loading waveform during the 750 cycles dictated the pace of the recovery during the extended free swelling period, as shorter inter-cycle recovery led to more persistent axial strain accumulation for up to five minutes. This work showcases the importance of fluid re-entry in resisting strain accumulation during cyclical compression.


Asunto(s)
Cartílago Articular , Humanos , Estrés Mecánico , Presión , Fenómenos Biomecánicos
19.
Nat Commun ; 15(1): 4170, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38755186

RESUMEN

Endothelial cells are a heterogeneous population with various organ-specific and conserved functions that are critical to organ development, function, and regeneration. Here we report a Sox17-Erg direct reprogramming approach that uses cardiac fibroblasts to create differentiated endothelial cells that demonstrate endothelial-like molecular and physiological functions in vitro and in vivo. Injection of these induced endothelial cells into myocardial infarct sites after injury results in improved vascular perfusion of the scar region. Furthermore, we use genomic analyses to illustrate that Sox17-Erg reprogramming instructs cardiac fibroblasts toward an arterial-like identity. This results in a more efficient direct conversion of fibroblasts into endothelial-like cells when compared to traditional Etv2-based reprogramming. Overall, this Sox17-Erg direct reprogramming strategy offers a robust tool to generate endothelial cells both in vitro and in vivo, and has the potential to be used in repairing injured tissue.


Asunto(s)
Reprogramación Celular , Células Endoteliales , Fibroblastos , Factores de Transcripción SOXF , Regulador Transcripcional ERG , Animales , Ratones , Diferenciación Celular , Reprogramación Celular/genética , Células Endoteliales/metabolismo , Células Endoteliales/citología , Fibroblastos/metabolismo , Fibroblastos/citología , Proteínas HMGB/metabolismo , Proteínas HMGB/genética , Ratones Endogámicos C57BL , Infarto del Miocardio/patología , Miocardio/citología , Miocardio/metabolismo , Factores de Transcripción SOXF/metabolismo , Factores de Transcripción SOXF/genética , Regulador Transcripcional ERG/genética , Regulador Transcripcional ERG/metabolismo
20.
bioRxiv ; 2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39229154

RESUMEN

Somatic activating mutations in PIK3CA are common drivers of vascular and lymphatic malformations. Despite common biophysical signatures of tissues susceptible to lesion formation, including compliant extracellular matrix and low rates of perfusion, lesions vary in clinical presentation from localized cystic dilatation to diffuse and infiltrative vascular dysplasia. The mechanisms driving the differences in disease severity and variability in clinical presentation and the role of the biophysical microenvironment in potentiating progression are poorly understood. Here, we investigate the role of hemodynamic forces and the biophysical microenvironment in the pathophysiology of vascular malformations, and we identify hemodynamic shear stress and defective endothelial cell mechanotransduction as key regulators of lesion progression. We found that constitutive PI3K activation impaired flow-mediated endothelial cell alignment and barrier function. We show that defective shear stress sensing in PIK3CA E542K endothelial cells is associated with reduced myosin light chain phosphorylation, junctional instability, and defective recruitment of vinculin to cell-cell junctions. Using 3D microfluidic models of the vasculature, we demonstrate that PIK3CA E542K microvessels apply reduced traction forces and are unaffected by flow interruption. We further found that draining transmural flow resulted in increased sprouting and invasion responses in PIK3CA E542K microvessels. Mechanistically, constitutive PI3K activation decreased cellular and nuclear elasticity resulting in defective cellular tensional homeostasis in endothelial cells which may underlie vascular dilation, tissue hyperplasia, and hypersprouting in PIK3CA-driven venous and lymphatic malformations. Together, these results suggest that defective nuclear mechanics, impaired cellular mechanotransduction, and maladaptive hemodynamic responses contribute to the development and progression of PIK3CA-driven vascular malformations.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA