Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.128
Filtrar
1.
Curr Top Dev Biol ; 159: 344-370, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38729681

RESUMEN

The development of the vascular system is crucial in supporting the growth and health of all other organs in the body, and vascular system dysfunction is the major cause of human morbidity and mortality. This chapter discusses three successive processes that govern vascular system development, starting with the differentiation of the primitive vascular system in early embryonic development, followed by its remodeling into a functional circulatory system composed of arteries and veins, and its final maturation and acquisition of an organ specific semi-permeable barrier that controls nutrient uptake into tissues and hence controls organ physiology. Along these steps, endothelial cells forming the inner lining of all blood vessels acquire extensive heterogeneity in terms of gene expression patterns and function, that we are only beginning to understand. These advances contribute to overall knowledge of vascular biology and are predicted to unlock the unprecedented therapeutic potential of the endothelium as an avenue for treatment of diseases associated with dysfunctional vasculature.


Asunto(s)
Remodelación Vascular , Humanos , Animales , Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/embriología , Neovascularización Fisiológica , Células Endoteliales/citología , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Diferenciación Celular , Desarrollo Embrionario , Endotelio Vascular/citología
3.
Development ; 149(3)2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34931661

RESUMEN

Endothelial cell migration and proliferation are essential for the establishment of a hierarchical organization of blood vessels and optimal distribution of blood. However, how these cellular processes are quantitatively coordinated to drive vascular network morphogenesis remains unknown. Here, using the zebrafish vasculature as a model system, we demonstrate that the balanced distribution of endothelial cells, as well as the resulting regularity of vessel calibre, is a result of cell migration from veins towards arteries and cell proliferation in veins. We identify the Wiskott-Aldrich Syndrome protein (WASp) as an important molecular regulator of this process and show that loss of coordinated migration from veins to arteries upon wasb depletion results in aberrant vessel morphology and the formation of persistent arteriovenous shunts. We demonstrate that WASp achieves its function through the coordination of junctional actin assembly and PECAM1 recruitment and provide evidence that this is conserved in humans. Overall, we demonstrate that functional vascular patterning in the zebrafish trunk is established through differential cell migration regulated by junctional actin, and that interruption of differential migration may represent a pathomechanism in vascular malformations.


Asunto(s)
Vasos Sanguíneos/crecimiento & desarrollo , Morfogénesis/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Proteína del Síndrome de Wiskott-Aldrich/genética , Actinas/genética , Animales , Arterias/crecimiento & desarrollo , Arterias/metabolismo , Movimiento Celular/genética , Proliferación Celular/genética , Células Endoteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Uniones Intercelulares/genética , Venas/crecimiento & desarrollo , Venas/metabolismo , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo
4.
Arterioscler Thromb Vasc Biol ; 41(12): 2974-2989, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34615374

RESUMEN

OBJECTIVE: Pathological angiogenesis is a hallmark of various diseases characterized by local hypoxia and inflammation. These disorders can be treated with inhibitors of angiogenesis, but current compounds display a variety of side effects and lose efficacy over time. This makes the identification of novel signaling pathways and pharmacological targets involved in angiogenesis a top priority. Approach and Results: Here, we show that inactivation of FAAH (fatty acid amide hydrolase), the enzyme responsible for degradation of the endocannabinoid anandamide, strongly impairs angiogenesis in vitro and in vivo. Both, the pharmacological FAAH inhibitor URB597 and anandamide induce downregulation of gene sets for cell cycle progression and DNA replication in endothelial cells. This is underscored by cell biological experiments, in which both compounds inhibit proliferation and migration and evoke cell cycle exit of endothelial cells. This prominent antiangiogenic effect is also of pathophysiological relevance in vivo, as laser-induced choroidal neovascularization in the eye of FAAH-/- mice is strongly reduced. CONCLUSIONS: Thus, elevation of endogenous anandamide levels by FAAH inhibition represents a novel antiangiogenic mechanism.


Asunto(s)
Amidohidrolasas/farmacocinética , Ácidos Araquidónicos/farmacología , Vasos Sanguíneos/efectos de los fármacos , Endocannabinoides/farmacología , Endotelio Vascular/crecimiento & desarrollo , Músculo Liso Vascular/efectos de los fármacos , Alcamidas Poliinsaturadas/farmacología , Animales , Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/patología , Agonistas de Receptores de Cannabinoides/farmacología , Bovinos , Línea Celular , Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Humanos , Ratones , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Neovascularización Patológica
5.
Int J Mol Sci ; 22(17)2021 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-34502102

RESUMEN

Every cell in the body requires oxygen for its functioning, in virtually every animal, and a tightly regulated system that balances oxygen supply and demand is therefore fundamental. The vascular network is one of the first systems to sense oxygen, and deprived oxygen (hypoxia) conditions automatically lead to a cascade of cellular signals that serve to circumvent the negative effects of hypoxia, such as angiogenesis associated with inflammation, tumor development, or vascular disorders. This vascular signaling is driven by central transcription factors, namely the hypoxia inducible factors (HIFs), which determine the expression of a growing number of genes in endothelial cells and pericytes. HIF functions are tightly regulated by oxygen sensors known as the HIF-prolyl hydroxylase domain proteins (PHDs), which are enzymes that hydroxylate HIFs for eventual proteasomal degradation. HIFs, as well as PHDs, represent attractive therapeutic targets under various pathological settings, including those involving vascular (dys)function. We focus on the characteristics and mechanisms by which vascular cells respond to hypoxia under a variety of conditions.


Asunto(s)
Proteínas Angiogénicas/metabolismo , Vasos Sanguíneos/metabolismo , Hipoxia de la Célula , Factor 1 Inducible por Hipoxia/metabolismo , Oxígeno/metabolismo , Proteínas Angiogénicas/genética , Animales , Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/fisiología , Redes Reguladoras de Genes , Humanos , Factor 1 Inducible por Hipoxia/genética , Neovascularización Fisiológica
6.
Sci Rep ; 11(1): 18251, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34521868

RESUMEN

Fluid flow shear stresses are strong regulators for directing the organization of vascular networks. Knowledge of structural and flow dynamics information within complex vasculature is essential for tuning the vascular organization within engineered tissues, by manipulating flows. However, reported investigations of vascular organization and their associated flow dynamics within complex vasculature over time are limited, due to limitations in the available physiological pre-clinical models, and the optical inaccessibility and aseptic nature of these models. Here, we developed laser speckle contrast imaging (LSCI) and side-stream dark field microscopy (SDF) systems to map the vascular organization, spatio-temporal blood flow fluctuations as well as erythrocytes movements within individual blood vessels of developing chick embryo, cultured within an artificial eggshell system. By combining imaging data and computational simulations, we estimated fluid flow shear stresses within multiscale vasculature of varying complexity. Furthermore, we demonstrated the LSCI compatibility with bioengineered perfusable muscle tissue constructs, fabricated via molding techniques. The presented application of LSCI and SDF on perfusable tissues enables us to study the flow perfusion effects in a non-invasive fashion. The gained knowledge can help to use fluid perfusion in order to tune and control multiscale vascular organization within engineered tissues.


Asunto(s)
Circulación Sanguínea , Vasos Sanguíneos/fisiología , Imagen Óptica/métodos , Ingeniería de Tejidos/métodos , Animales , Vasos Sanguíneos/diagnóstico por imagen , Vasos Sanguíneos/crecimiento & desarrollo , Embrión de Pollo , Eritrocitos/fisiología , Imagen Multimodal/métodos , Músculos/irrigación sanguínea , Músculos/diagnóstico por imagen , Neovascularización Fisiológica
7.
Am J Pathol ; 191(12): 2245-2264, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34563512

RESUMEN

Whether alterations in the microtubule cytoskeleton affect the ability of endothelial cells (ECs) to sprout and form branching networks of tubes was investigated in this study. Bioassays of human EC tubulogenesis, where both sprouting behavior and lumen formation can be rigorously evaluated, were used to demonstrate that addition of the microtubule-stabilizing drugs, paclitaxel, docetaxel, ixabepilone, and epothilone B, completely interferes with EC tip cells and sprouting behavior, while allowing for EC lumen formation. In bioassays mimicking vasculogenesis using single or aggregated ECs, these drugs induce ring-like lumens from single cells or cyst-like spherical lumens from multicellular aggregates with no evidence of EC sprouting behavior. Remarkably, treatment of these cultures with a low dose of the microtubule-destabilizing drug, vinblastine, led to an identical result, with complete blockade of EC sprouting, but allowing for EC lumen formation. Administration of paclitaxel in vivo markedly interfered with angiogenic sprouting behavior in developing mouse retina, providing corroboration. These findings reveal novel biological activities for pharmacologic agents that are widely utilized in multidrug chemotherapeutic regimens for the treatment of human malignant cancers. Overall, this work demonstrates that manipulation of microtubule stability selectively interferes with the ability of ECs to sprout, a necessary step to initiate and form branched capillary tube networks.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Neovascularización Patológica/tratamiento farmacológico , Paclitaxel/farmacología , Animales , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/crecimiento & desarrollo , Células Cultivadas , Docetaxel/farmacología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/crecimiento & desarrollo , Epotilonas/farmacología , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Ratones , Ratones Endogámicos C57BL , Morfogénesis/efectos de los fármacos , Neovascularización Patológica/patología , Neovascularización Fisiológica/efectos de los fármacos , Paclitaxel/análogos & derivados
8.
Nat Commun ; 12(1): 4872, 2021 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-34381052

RESUMEN

The Netrin-1 receptor UNC5B is an axon guidance regulator that is also expressed in endothelial cells (ECs), where it finely controls developmental and tumor angiogenesis. In the absence of Netrin-1, UNC5B induces apoptosis that is blocked upon Netrin-1 binding. Here, we identify an UNC5B splicing isoform (called UNC5B-Δ8) expressed exclusively by ECs and generated through exon skipping by NOVA2, an alternative splicing factor regulating vascular development. We show that UNC5B-Δ8 is a constitutively pro-apoptotic splicing isoform insensitive to Netrin-1 and required for specific blood vessel development in an apoptosis-dependent manner. Like NOVA2, UNC5B-Δ8 is aberrantly expressed in colon cancer vasculature where its expression correlates with tumor angiogenesis and poor patient outcome. Collectively, our data identify a mechanism controlling UNC5B's necessary apoptotic function in ECs and suggest that the NOVA2/UNC5B circuit represents a post-transcriptional pathway regulating angiogenesis.


Asunto(s)
Apoptosis , Vasos Sanguíneos/crecimiento & desarrollo , Receptores de Netrina/metabolismo , Isoformas de ARN/metabolismo , Empalme Alternativo , Animales , Neoplasias del Colon/irrigación sanguínea , Neoplasias del Colon/metabolismo , Células Endoteliales , Humanos , Morfogénesis , Neovascularización Patológica/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Receptores de Netrina/genética , Netrina-1/metabolismo , Antígeno Ventral Neuro-Oncológico , Isoformas de ARN/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Análisis de Supervivencia , Pez Cebra
9.
Int J Mol Sci ; 22(11)2021 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-34204949

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is one of the most symptomatic progressive fibrotic lung diseases, in which patients have an extremely poor prognosis. Therefore, understanding the precise molecular mechanisms underlying pulmonary fibrosis is necessary for the development of new therapeutic options. Stress-activated protein kinases (SAPKs), c-Jun N-terminal kinase (JNK), and p38 mitogen-activated protein kinase (p38) are ubiquitously expressed in various types of cells and activated in response to cellular environmental stresses, including inflammatory and apoptotic stimuli. Type II alveolar epithelial cells, fibroblasts, and macrophages are known to participate in the progression of pulmonary fibrosis. SAPKs can control fibrogenesis by regulating the cellular processes and molecular functions in various types of lung cells (including cells of the epithelium, interstitial connective tissue, blood vessels, and hematopoietic and lymphoid tissue), all aspects of which remain to be elucidated. We recently reported that the stepwise elevation of intrinsic p38 signaling in the lungs is correlated with a worsening severity of bleomycin-induced fibrosis, indicating an importance of this pathway in the progression of pulmonary fibrosis. In addition, a transcriptome analysis of RNA-sequencing data from this unique model demonstrated that several lines of mechanisms are involved in the pathogenesis of pulmonary fibrosis, which provides a basis for further studies. Here, we review the accumulating evidence for the spatial and temporal roles of SAPKs in pulmonary fibrosis.


Asunto(s)
Fibrosis Pulmonar Idiopática/genética , Proteínas Quinasas JNK Activadas por Mitógenos/genética , MAP Quinasa Quinasa 4/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Vasos Sanguíneos/enzimología , Vasos Sanguíneos/crecimiento & desarrollo , Fibroblastos/enzimología , Humanos , Fibrosis Pulmonar Idiopática/enzimología , Fibrosis Pulmonar Idiopática/patología , Pulmón/embriología , Pulmón/patología , Sistema de Señalización de MAP Quinasas/genética , Macrófagos/enzimología
10.
Methods Mol Biol ; 2319: 93-104, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34331247

RESUMEN

Lightsheet microscopy is a form of fluorescence microscopy that can be used to visualize specimen with high resolution, a large depth-of-field, and minimal photodamage and photobleaching as compared to traditional confocal microscopy. As this technology becomes much more readily available, it will be useful in revealing new findings in the cardiovascular development field that may be hidden or difficult to image. In this manuscript, we describe an approach for mounting and culturing postimplantation mouse embryos to visualize blood vessel development with a lightsheet microscope.


Asunto(s)
Angiografía/métodos , Vasos Sanguíneos/diagnóstico por imagen , Técnicas de Cultivo/métodos , Embrión de Mamíferos/diagnóstico por imagen , Desarrollo Embrionario , Microscopía Fluorescente/métodos , Neovascularización Fisiológica , Animales , Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/metabolismo , Medios de Cultivo/química , Disección/métodos , Embrión de Mamíferos/irrigación sanguínea , Ratones , Ratones Transgénicos , Microscopía Confocal
11.
PLoS Comput Biol ; 17(6): e1009094, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34181657

RESUMEN

Angiogenesis is the process by which blood vessels form from pre-existing vessels. It plays a key role in many biological processes, including embryonic development and wound healing, and contributes to many diseases including cancer and rheumatoid arthritis. The structure of the resulting vessel networks determines their ability to deliver nutrients and remove waste products from biological tissues. Here we simulate the Anderson-Chaplain model of angiogenesis at different parameter values and quantify the vessel architectures of the resulting synthetic data. Specifically, we propose a topological data analysis (TDA) pipeline for systematic analysis of the model. TDA is a vibrant and relatively new field of computational mathematics for studying the shape of data. We compute topological and standard descriptors of model simulations generated by different parameter values. We show that TDA of model simulation data stratifies parameter space into regions with similar vessel morphology. The methodologies proposed here are widely applicable to other synthetic and experimental data including wound healing, development, and plant biology.


Asunto(s)
Modelos Cardiovasculares , Neovascularización Patológica , Neovascularización Fisiológica , Algoritmos , Animales , Vasos Sanguíneos/anatomía & histología , Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/fisiología , Quimiotaxis , Biología Computacional , Simulación por Computador , Humanos , Neoplasias/irrigación sanguínea , Análisis Espacio-Temporal
12.
Int J Comput Assist Radiol Surg ; 16(8): 1243-1254, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34125391

RESUMEN

PURPOSE: Intravascular ultrasound (IVUS) imaging is crucial for planning and performing percutaneous coronary interventions. Automatic segmentation of lumen and vessel wall in IVUS images can thus help streamlining the clinical workflow. State-of-the-art results in image segmentation are achieved with data-driven methods like convolutional neural networks (CNNs). These need large amounts of training data to perform sufficiently well but medical image datasets are often rather small. A possibility to overcome this problem is exploiting alternative network architectures like capsule networks. METHODS: We systematically investigated different capsule network architecture variants and optimized the performance on IVUS image segmentation. We then compared our capsule network with corresponding CNNs under varying amounts of training images and network parameters. RESULTS: Contrary to previous works, our capsule network performs best when doubling the number of capsule types after each downsampling stage, analogous to typical increase rates of feature maps in CNNs. Maximum improvements compared to the baseline CNNs are 20.6% in terms of the Dice coefficient and 87.2% in terms of the average Hausdorff distance. CONCLUSION: Capsule networks are promising candidates when it comes to segmentation of small IVUS image datasets. We therefore assume that this also holds for ultrasound images in general. A reasonable next step would be the investigation of capsule networks for few- or even single-shot learning tasks.


Asunto(s)
Vasos Sanguíneos/crecimiento & desarrollo , Procesamiento de Imagen Asistido por Computador/métodos , Redes Neurales de la Computación , Ultrasonografía Intervencional/métodos , Humanos
13.
Dev Cell ; 56(13): 1848-1860, 2021 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-34146467

RESUMEN

In mammals, hematopoietic stem cells (HSCs) engage in hematopoiesis throughout adult life within the bone marrow, where they produce the mature cells necessary to maintain blood cell counts and immune function. In the bone marrow and spleen, HSCs are sustained in perivascular niches (microenvironments) associated with sinusoidal blood vessels-specialized veins found only in hematopoietic tissues. Endothelial cells and perivascular leptin receptor+ stromal cells produce the known factors required to maintain HSCs and many restricted progenitors in the bone marrow. Various other cells synthesize factors that maintain other restricted progenitors or modulate HSC or niche function. Recent studies identified new markers that resolve some of the heterogeneity among stromal cells and refine the localization of restricted progenitor niches. Other recent studies identified ways in which niches regulate HSC function and hematopoiesis beyond growth factors. We summarize the current understanding of hematopoietic niches, review recent progress, and identify important unresolved questions.


Asunto(s)
Médula Ósea/metabolismo , Hematopoyesis/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Células Madre/citología , Vasos Sanguíneos/citología , Vasos Sanguíneos/crecimiento & desarrollo , Médula Ósea/crecimiento & desarrollo , Células Endoteliales/metabolismo , Humanos , Receptores de Leptina/genética , Bazo/citología , Bazo/metabolismo , Nicho de Células Madre/genética , Células Madre/metabolismo
14.
Dev Dyn ; 250(12): 1717-1738, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34115420

RESUMEN

BACKGROUND: A significant challenge facing tissue engineering is the fabrication of vasculature constructs which contains vascularized tissue constructs to recapitulate viable, complex and functional organs or tissues, and free-standing vascular structures potentially providing clinical applications in the future. Three-dimensional (3D) bioprinting has emerged as a promising technology, possessing a number of merits that other conventional biofabrication methods do not have. Over the last decade, 3D bioprinting has contributed a variety of techniques and strategies to generate both vascularized tissue constructs and free-standing vascular structures. RESULTS: This review focuses on different strategies to print two kinds of vasculature constructs, namely vascularized tissue constructs and vessel-like tubular structures, highlighting the feasibility and shortcoming of the current methods for vasculature constructs fabrication. Generally, both direct printing and indirect printing can be employed in vascularized tissue engineering. Direct printing allows for structural fabrication with synchronous cell seeding, while indirect printing is more effective in generating complex architecture. During the fabrication process, 3D bioprinting techniques including extrusion bioprinting, inkjet bioprinting and light-assisted bioprinting should be selectively implemented to exert advantages and obtain the desirable tissue structure. Also, appropriate cells and biomaterials matter a lot to match various bioprinting techniques and thus achieve successful fabrication of specific vasculature constructs. CONCLUSION: The 3D bioprinting has been developed to help provide various fabrication techniques, devoting to producing structurally stable, physiologically relevant, and biologically appealing constructs. However, although the optimization of biomaterials and innovation of printing strategies may improve the fabricated vessel-like structures, 3D bioprinting is still in the infant period and has a great gap between in vitro trials and in vivo applications. The article reviews the present achievement of 3D bioprinting in generating vasculature constructs and also provides perspectives on future directions of advanced vasculature constructs fabrication.


Asunto(s)
Bioimpresión , Vasos Sanguíneos/citología , Impresión Tridimensional , Ingeniería de Tejidos/tendencias , Animales , Bioimpresión/métodos , Bioimpresión/tendencias , Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/fisiología , Humanos , Impresión Tridimensional/tendencias , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Andamios del Tejido/tendencias
15.
J Cell Mol Med ; 25(14): 6664-6678, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34117708

RESUMEN

Synovial macrophage polarization and inflammation are essential for osteoarthritis (OA) development, yet the molecular mechanisms and regulation responsible for the pathogenesis are still poorly understood. Here, we report that pseudolaric acid B (PAB) attenuated articular cartilage degeneration and synovitis during OA. PAB, a diterpene acid, specifically inhibited NF-κB signalling and reduced the production of pro-inflammatory cytokines, which further decreased M1 polarization and vessel formation. We further provide in vivo and in vitro evidences that PAB suppressed NF-κB signalling by stabilizing PPARγ. Using PPARγ antagonist could abolish anti-inflammatory effect of PAB and rescue the activation of NF-κB signalling during OA. Our findings identify a previously unrecognized role of PAB in the regulation of OA and provide mechanisms by which PAB regulates NF-κB signalling through PPARγ, which further suggest targeting synovial inflammation or inhibiting vessel formation at early stage could be an effective preventive strategy for OA.


Asunto(s)
Diterpenos/farmacología , Osteoartritis/tratamiento farmacológico , PPAR gamma/genética , Sinovitis/tratamiento farmacológico , Animales , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/crecimiento & desarrollo , Cartílago Articular/efectos de los fármacos , Cartílago Articular/crecimiento & desarrollo , Cartílago Articular/patología , Condrocitos/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/parasitología , Ratones , FN-kappa B/genética , Osteoartritis/genética , Osteoartritis/patología , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos , Sinovitis/genética , Sinovitis/patología , Factor de Transcripción ReIA/genética
16.
Dev Biol ; 477: 64-69, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34019880

RESUMEN

Cdc42, a Rho family low molecular weight G protein, has important roles in various cell functions, including cytoskeletal rearrangement, cell adhesion and cell proliferation and differentiation. To investigate the involvement of Cdc42 in the activities of vascular endothelial cells, we generated Cdc42 conditional knockout mice in which Cdc42 was time -specifically deficient in vascular endothelial cells (Cdc42 â€‹fl/fl; VE-Cad CreERT: Cdc42 cKO). When the Cdc42 gene was deleted after birth, Cdc42 cKO mice were smaller than the control mice, and died between postnatal day 8 (P8) and P10. Necropsy findings confirmed that these mice had various pathological aberrances in the vessels of most organs, such as blood flow congestion and blood cell invasion. Electron microscopic observations also revealed that capillary endothelial cells were detached from the basement membrane as well as phagocytosis of dead endothelial cells induced by macrophages. Moreover, vascular sprouting from aortic rings induced by VEGF-A was diminished in samples from the Cdc42 cKO mice because of an endothelial cell proliferation defect. These results suggest that Cdc42 in vascular endothelial cells has important roles in blood vessel formation after birth.


Asunto(s)
Vasos Sanguíneos/crecimiento & desarrollo , Células Endoteliales/fisiología , Neovascularización Fisiológica/fisiología , Proteína de Unión al GTP cdc42/fisiología , Animales , Ratones Noqueados
17.
Int J Mol Sci ; 22(8)2021 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-33919955

RESUMEN

Coculture systems employing adipose tissue-derived mesenchymal stromal/stem cells (ASC) and endothelial cells (EC) represent a widely used technique to model vascularization. Within this system, cell-cell communication is crucial for the achievement of functional vascular network formation. Extracellular vesicles (EVs) have recently emerged as key players in cell communication by transferring bioactive molecules between cells. In this study we aimed to address the role of EVs in ASC/EC cocultures by discriminating between cells, which have received functional EV cargo from cells that have not. Therefore, we employed the Cre-loxP system, which is based on donor cells expressing the Cre recombinase, whose mRNA was previously shown to be packaged into EVs and reporter cells containing a construct of floxed dsRed upstream of the eGFP coding sequence. The evaluation of Cre induced color switch in the reporter system via EVs indicated that there is no EV-mediated RNA transmission either between EC themselves or EC and ASC. However, since Cre mRNA was not found present in EVs, it remains unclear if Cre mRNA is generally not packaged into EVs or if EVs are not taken up by the utilized cell types. Our data indicate that this technique may not be applicable to evaluate EV-mediated cell-to-cell communication in an in vitro setting using EC and ASC. Further investigations will require a functional system showing efficient and specific loading of Cre mRNA or protein into EVs.


Asunto(s)
Vesículas Extracelulares/genética , Integrasas/genética , Células Madre Mesenquimatosas/metabolismo , MicroARNs/genética , Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/metabolismo , Comunicación Celular/genética , Técnicas de Cocultivo , Células Endoteliales/metabolismo , Humanos , Células Madre Mesenquimatosas/citología , ARN Mensajero/genética
18.
Int J Mol Sci ; 22(8)2021 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-33920100

RESUMEN

Chitinase 3-like 1 (CHI3L1) is an enzymatically inactive mammalian chitinase that is associated with tumor inflammation. Previous research indicated that CHI3L1 is able to interact with different extracellular matrix components, such as heparan sulfate. In the present work, we investigated whether the interaction of CHI3L1 with the extracellular matrix of melanoma cells can trigger an inflammatory activation of endothelial cells. The analysis of the melanoma cell secretome indicated that CHI3L1 increases the abundance of various cytokines, such as CC-chemokine ligand 2 (CCL2), and growth factors, such as vascular endothelial growth factor A (VEGF-A). Using a solid-phase binding assay, we found that heparan sulfate-bound VEGF-A and CCL2 were displaced by recombinant CHI3L1 in a dose-dependent manner. Microfluidic experiments indicated that the CHI3L1 altered melanoma cell secretome promoted immune cell recruitment to the vascular endothelium. In line with the elevated VEGF-A levels, CHI3L1 was also able to promote angiogenesis through the release of extracellular matrix-bound pro-angiogenic factors. In conclusion, we showed that CHI3L1 is able to affect the tumor cell secretome, which in turn can regulate immune cell recruitment and blood vessel formation. Accordingly, our data suggest that the molecular targeting of CHI3L1 in the course of cancer immunotherapies can tune patients' response and antitumoral inflammation.


Asunto(s)
Quimiocina CCL2/genética , Proteína 1 Similar a Quitinasa-3/genética , Melanoma/genética , Neovascularización Patológica/genética , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/inmunología , Vasos Sanguíneos/patología , Línea Celular Tumoral , Células Endoteliales/inmunología , Células Endoteliales/patología , Endotelio Vascular/crecimiento & desarrollo , Endotelio Vascular/inmunología , Endotelio Vascular/patología , Matriz Extracelular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glicosaminoglicanos/farmacología , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Melanoma/inmunología , Melanoma/patología , Técnicas Analíticas Microfluídicas , Neovascularización Patológica/inmunología , Neovascularización Patológica/patología , Unión Proteica/genética , Unión Proteica/inmunología
19.
Nat Cell Biol ; 23(4): 322-329, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33837285

RESUMEN

De novo blood vessel formation occurs through coalescence of endothelial cells (ECs) into a cord-like structure, followed by lumenization either through cell-1-3 or cord-hollowing4-7. Vessels generated in this manner are restricted in diameter to one or two ECs, and these models fail to explain how vasculogenesis can form large-diameter vessels. Here, we describe a model for large vessel formation that does not require a cord-like structure or a hollowing step. In this model, ECs coalesce into a network of struts in the future lumen of the vessel, a process dependent upon bone morphogenetic protein signalling. The vessel wall forms around this network and consists initially of only a few patches of ECs. To withstand external forces and to maintain the shape of the vessel, strut formation traps erythrocytes into compartments to form a rigid structure. Struts gradually prune and ECs from struts migrate into and become part of the vessel wall. Experimental severing of struts resulted in vessel collapse, disturbed blood flow and remodelling defects, demonstrating that struts enable the patency of large vessels during their formation.


Asunto(s)
Vasos Sanguíneos/crecimiento & desarrollo , Células Endoteliales/fisiología , Morfogénesis/genética , Neovascularización Fisiológica/genética , Vasos Sanguíneos/metabolismo , Células Endoteliales/metabolismo , Eritrocitos/metabolismo , Eritrocitos/patología , Humanos
20.
Biochem Biophys Res Commun ; 557: 8-13, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-33857842

RESUMEN

Precise quantification of vascular developments in Zebrafish requires continuous in-vivo 3D imaging. Here we employed a bi-directional light-sheet illumination microscope to characterize the development process of Zebrafish's intersegmental vessels. A Virtual Reality-based method was used to measure the lengths of intersegmental vessels (ISVs). The quantified growth rates of typical ISVs can be plotted, and unusual growth of some specific vessels was also observed.


Asunto(s)
Vasos Sanguíneos/embriología , Embrión no Mamífero/irrigación sanguínea , Microscopía/instrumentación , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente , Vasos Sanguíneos/crecimiento & desarrollo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Imagenología Tridimensional/métodos , Iluminación , Microscopía/métodos , Neovascularización Fisiológica , Pez Cebra/crecimiento & desarrollo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...