Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 898
Filtrar
1.
Curr Med Sci ; 44(3): 578-588, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38853191

RESUMEN

OBJECTIVE: Brain microvascular endothelial cells (BMECs) were found to shift from their usually inactive state to an active state in ischemic stroke (IS) and cause neuronal damage. Ginsenoside Rb1 (GRb1), a component derived from medicinal plants, is known for its pharmacological benefits in IS, but its protective effects on BMECs have yet to be explored. This study aimed to investigate the potential protective effects of GRb1 on BMECs. METHODS: An in vitro oxygen-glucose deprivation/reperfusion (OGD/R) model was established to mimic ischemia-reperfusion (I/R) injury. Bulk RNA-sequencing data were analyzed by using the Human Autophagy Database and various bioinformatic tools, including gene set enrichment analysis (GSEA), Gene Ontology (GO) classification and enrichment analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, protein-protein interaction network analysis, and molecular docking. Experimental validation was also performed to ensure the reliability of our findings. RESULTS: Rb1 had a protective effect on BMECs subjected to OGD/R injury. Specifically, GRb1 was found to modulate the interplay between oxidative stress, apoptosis, and autophagy in BMECs. Key targets such as sequestosome 1 (SQSTM1/p62), autophagy related 5 (ATG5), and hypoxia-inducible factor 1-alpha (HIF-1α) were identified, highlighting their potential roles in mediating the protective effects of GRb1 against IS-induced damage. CONCLUSION: GRbl protects BMECs against OGD/R injury by influencing oxidative stress, apoptosis, and autophagy. The identification of SQSTM1/p62, ATG5, and HIF-1α as promising targets further supports the potential of GRb1 as a therapeutic agent for IS, providing a foundation for future research into its mechanisms and applications in IS treatment.


Asunto(s)
Apoptosis , Autofagia , Células Endoteliales , Ginsenósidos , Estrés Oxidativo , Ginsenósidos/farmacología , Estrés Oxidativo/efectos de los fármacos , Autofagia/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Apoptosis/efectos de los fármacos , Humanos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Simulación del Acoplamiento Molecular , Mapas de Interacción de Proteínas/efectos de los fármacos , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , Microvasos/efectos de los fármacos , Microvasos/citología , Microvasos/metabolismo , Biología Computacional/métodos , Glucosa/metabolismo
2.
Microcirculation ; 31(5): e12859, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38818977

RESUMEN

OBJECTIVE: The endothelium regulates crucial aspects of vascular function, including hemostasis, vasomotor tone, proliferation, immune cell adhesion, and microvascular permeability. Endothelial cells (ECs), especially in arterioles, are pivotal for flow distribution and peripheral resistance regulation. Investigating vascular endothelium physiology, particularly in microvascular ECs, demands precise isolation and culturing techniques. METHODS: Freshly isolated ECs are vital for examining protein expression, ion channel behavior, and calcium dynamics. Establishing primary endothelial cell cultures is crucial for unraveling vascular functions and understanding intact microvessel endothelium roles. Despite the significance, detailed protocols and comparisons with intact vessels are scarce in microvascular research. We developed a reproducible method to isolate microvascular ECs, assessing substrate influence by cultivating cells on fibronectin and gelatin matrix gels. This comparative approach enhances our understanding of microvascular endothelial cell biology. RESULTS: Microvascular mesenteric ECs expressed key markers (VE-cadherin and eNOS) in both matrix gels, confirming cell culture purity. Under uncoated conditions, ECs were undetected, whereas proteins linked to smooth muscle cells and fibroblasts were evident. Examining endothelial cell (EC) physiological dynamics on distinct matrix substrates revealed comparable cell length, shape, and Ca2+ elevations in both male and female ECs on gelatin and fibronectin matrix gels. Gelatin-cultured ECs exhibited analogous membrane potential responses to acetylcholine (ACh) or adenosine triphosphate (ATP), contrasting with their fibronectin-cultured counterparts. In the absence of stimulation, fibronectin-cultured ECs displayed a more depolarized resting membrane potential than gelatin-cultured ECs. CONCLUSIONS: Gelatin-cultured ECs demonstrated electrical behaviors akin to intact endothelium from mouse mesenteric arteries, thus advancing our understanding of endothelial cell behavior within diverse microenvironments.


Asunto(s)
Células Endoteliales , Gelatina , Microvasos , Óxido Nítrico Sintasa de Tipo III , Animales , Células Endoteliales/metabolismo , Células Endoteliales/citología , Ratones , Femenino , Masculino , Microvasos/citología , Microvasos/metabolismo , Microvasos/fisiología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Células Cultivadas , Fibronectinas/metabolismo , Fibronectinas/farmacología , Geles , Antígenos CD/metabolismo , Cadherinas/metabolismo , Cultivo Primario de Células , Endotelio Vascular/metabolismo , Endotelio Vascular/citología
3.
J Dent Res ; 103(6): 652-661, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38716736

RESUMEN

The process of neovascularization during cell-based pulp regeneration is difficult to study. Here we developed a tube model that simulates root canal space and allows direct visualization of the vascularization process in vitro. Endothelial-like cells (ECs) derived from guiding human dental pulp stem cells (DPSCs) into expressing endothelial cell markers CD144, vWF, VEGFR1, and VEGFR2 were used. Human microvascular endothelial cells (hMVECs) were used as a positive control. DPSC-ECs formed tubules on Matrigel similar to hMVECs. Cells were mixed in fibrinogen/thrombin or mouse blood and seeded into wells of 96-well plates or injected into a tapered plastic tube (14 mm in length and 1 or 2 mm diameter of the apex opening) with the larger end sealed with MTA to simulate root canal space. Cells/gels in wells or tubes were incubated for various times in vitro and observed under the microscope for morphological changes. Samples were then fixed and processed for histological analysis to determine vessel formation. Vessel-like networks were observed in culture from 1 to 3 d after cell seeding. Cells/gels in 96-well plates were maintained up to 25 d. Histologically, both hMVECs and DPSC-ECs in 96-well plates or tubes showed intracellular vacuole formation. Some cells showed merged large vacuoles indicating the lumenization. Tubular structures were also observed resembling blood vessels. Cells appeared healthy throughout the tube except some samples (1 mm apical diameter) in the coronal third. Histological analysis also showed pulp-like soft tissue throughout the tube samples with vascular-like structures. hMVECs formed larger vascular lumen size than DPSC-ECs while the latter tended to have more lumen and tubular structure counts. We conclude that DPSC-ECs can form vascular structures and sustained in the 3-dimensional fibrin gel system in vitro. The tube model appears to be a proper and simple system simulating the root canal space for vascular formation and pulp regeneration studies.


Asunto(s)
Pulpa Dental , Combinación de Medicamentos , Células Endoteliales , Neovascularización Fisiológica , Proteoglicanos , Regeneración , Células Madre , Pulpa Dental/citología , Pulpa Dental/irrigación sanguínea , Pulpa Dental/fisiología , Neovascularización Fisiológica/fisiología , Animales , Ratones , Humanos , Regeneración/fisiología , Células Endoteliales/fisiología , Células Madre/fisiología , Colágeno , Técnicas de Cultivo de Célula , Laminina , Factor de von Willebrand/análisis , Receptor 2 de Factores de Crecimiento Endotelial Vascular , Fibrinógeno , Cavidad Pulpar , Compuestos de Calcio , Compuestos de Aluminio , Materiales de Obturación del Conducto Radicular , Microvasos/citología , Células Cultivadas , Óxidos , Silicatos , Antígeno CD146
4.
Tissue Eng Part C Methods ; 30(5): 229-237, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38568845

RESUMEN

Modeling organ-blood barriers through the inclusion of microvessel networks within in vitro tissue models could lead to more physiologically accurate results, especially since organ-blood barriers are crucial to the normal function, drug transport, and disease states of vascularized organs. Microvessel networks are difficult to form, since they push the practical limits of most fabrication methods, and it is difficult to coax vascular cells to self-assemble into structures larger than capillaries. Here, we present a method for rapidly forming networks of microvessel-like structures using sacrificial alginate structures. Specifically, we encapsulated endothelial cells within short alginate threads, and then embedded them in collagen gel. Following enzymatic degradation of the alginate, the collagen gel contained a network of hollow channels seeded with cells, all surrounding a perfusable central channel. This method uses a 3D-printed coaxial extruder and syringe pumps to generate short threads in a way that is repeatable and easily transferrable to other labs. The cell-laden, sacrificial alginate threads can be frozen after fabrication and thawed before embedding without significant loss of cell viability. The ability to freeze the threads enables future scale-up and ease of use. Within millifluidic devices that restrict access to media, the threads enhance cell survival under static conditions. These results indicate the potential for use of this method in a range of tissue engineering applications.


Asunto(s)
Alginatos , Microvasos , Ingeniería de Tejidos , Alginatos/química , Microvasos/citología , Humanos , Ingeniería de Tejidos/métodos , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Andamios del Tejido/química , Células Endoteliales/citología , Células Endoteliales/metabolismo , Ácido Glucurónico/química , Ácidos Hexurónicos/química , Supervivencia Celular , Animales , Colágeno/química
5.
STAR Protoc ; 5(2): 102950, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38483899

RESUMEN

Organ-on-a-chip technologies enable the fabrication of endothelial tissues, so-called microvessels (MVs), which emulate the endothelial barrier function in healthy or disease conditions. In this protocol, we describe the fabrication of perfusable open-chamber style MVs embedded in collagen gels. We then report a simple technology to characterize the MV barrier properties in static or under pressure based on fluorescence confocal imaging. Finally, we provide quantification techniques that enable us to infer the structure of MV paracellular pores. For complete details on the use and execution of this protocol, please refer to Cacheux et al.1.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana , Dispositivos Laboratorio en un Chip , Microvasos , Humanos , Microvasos/citología , Microscopía Confocal/métodos
6.
Adv Healthc Mater ; 13(12): e2303928, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38291861

RESUMEN

Chirality is an intrinsic cellular property that describes cell polarization biases along the left-right axis, apicobasal axis, or front-rear axes. Cell chirality plays a significant role in the arrangement of organs in the body as well as in the orientation of organelles, cytoskeletons, and cells. Vascular networks within the endometrium, the mucosal inner lining of the uterus, commonly display spiral architectures that rapidly form across the menstrual cycle. Herein, the role of endometrial-relevant extracellular matrix stiffness, composition, and soluble signals on endometrial endothelial cell chirality is systematically examined using a high-throughput microarray. Endometrial endothelial cells display marked patterns of chirality as individual cells and as cohorts in response to substrate stiffness and environmental cues. Vascular networks formed from endometrial endothelial cells also display shifts in chirality as a function of exogenous hormones. Changes in cellular-scale chirality correlate with changes in vascular network parameters, suggesting a critical role for cellular chirality in directing endometrial vessel network organization.


Asunto(s)
Endometrio , Células Endoteliales , Endometrio/citología , Endometrio/irrigación sanguínea , Endometrio/metabolismo , Humanos , Femenino , Células Endoteliales/citología , Células Endoteliales/metabolismo , Polaridad Celular/fisiología , Microvasos/citología , Microvasos/fisiología , Matriz Extracelular/metabolismo , Células Cultivadas
7.
Arch Biochem Biophys ; 720: 109173, 2022 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-35300940

RESUMEN

The effect of Esculetin on pyroptosis and its possible mechanism in endothelium were explored. 10 µg/mL LPS and 0.5 mM ATP were used to stimulate the rat intestinal microvascular endothelial cells. Then add different concentrations of Esculetin (20µM, 40 µM) to the culture medium containing LPS and ATP culturing for 24 h. The expression of p-NF-κB p65, NF-κB p65, I-κB, p-I-κB, NLRP3, ASC, caspase-1, and gasdermin-D were detected by Western blot, and the release level of IL-18 and IL-1ß were measured by ELISA. The NLRP3 inhibitor MCC950 was used at the concentration of 10 µM for 4 h to disentangle the potential mechanism of the influence of Esculetin on pyroptosis. In our experiments, the expression of gasdermin-d and important proteins of NF-κB and NLRP3 signaling pathways were inhibited by Esculetin. Besides, Esculetin also attenuated the morphological changes like swelling rupture and pores on the membrane caused by pyroptosis thereby protecting cells from being damaged by pyroptosis. Combining with the effect of Esculetin on proteins above and its protective effect on cell morphology, we believe that Esculetin has an anti-pyroptosis effect. The inhibiting pyroptosis effects mentioned above are similar to MCC950, which means the anti-pyroptosis effects of Esculetin are associated with the NLRP3 signaling pathway. In conclusion, Esculetin inhibits the pyroptosis of microvascular endothelial cells through the NF-κB/NLFP3 signaling pathway and is expected to be conducive in treating pyroptosis-related diseases.


Asunto(s)
Células Endoteliales , Microvasos , FN-kappa B , Piroptosis , Umbeliferonas , Adenosina Trifosfato , Animales , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Lipopolisacáridos/farmacología , Microvasos/citología , Microvasos/efectos de los fármacos , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Piroptosis/efectos de los fármacos , Ratas , Transducción de Señal , Umbeliferonas/farmacología
8.
Bioengineered ; 13(2): 4385-4396, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35139776

RESUMEN

As one of the most frequently prescribed antidiabetic drugs, metformin can lower glucose levels, improve insulin resistance manage body weight. However, the effect of metformin on islet microcirculation remains unclear. In the present study, to explore the effect of metformin on islet endothelial cells and investigated the underlying mechanism, we assessed the effects of metformin on islet endothelial cell survival, proliferation, oxidative stress and apoptosis. Our results suggest that metformin stimulates the proliferation of pancreatic islet endothelial cells and inhibits the apoptosis and oxidative stress caused by high glucose levels. By activating farnesoid X receptor (FXR), metformin increases the expression of vascular endothelial growth factor-A (VEGF-A) and endothelial nitric oxide synthase (eNOS), improves the production of nitric oxide (NO) and decreases the production of ROS. After the inhibition of FXR or VEGF-A, all of the effects disappeared. Thus, metformin appears to regulate islet microvascular endothelial cell (IMEC) proliferation, apoptosis and oxidative stress by activating the FXR/VEGF-A/eNOS pathway. These findings provide a new mechanism underlying the islet-protective effect of metformin.


Asunto(s)
Glucosa/efectos adversos , Islotes Pancreáticos , Metformina/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Endotelio Vascular/citología , Islotes Pancreáticos/irrigación sanguínea , Islotes Pancreáticos/citología , Islotes Pancreáticos/efectos de los fármacos , Ratones , Microvasos/citología , Estrés Oxidativo/efectos de los fármacos
9.
Biochem Pharmacol ; 195: 114745, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34454930

RESUMEN

The repair of vascular endothelial cell dysfunction is an encouraging approach for the treatment of vascular complications associated with diabetes. It has been demonstrated that members of C1q/tumor necrosis factor-related protein (CTRP) family may improve endothelial function. Nevertheless, the protective properties of CTRPs in diabetic microvascular complications continue to be mostly unknown. Here, we demonstrate that the C1q-like globular domain of CTRP3, CTRP5, and CTRP9 (gCTRP3, 5, 9) exerted a vasorelaxant effect on the microvasculature, of which gCTRP3 was the most powerful one. In a murine model of type 2 diabetes mellitus, serum gCTRP3 level and endothelial function decreased markedly compared with controls. Two weeks of gCTRP3 treatment (0.5 µg/g/d) enhanced endothelium-dependent relaxation in microvessels, increased nitric oxide (NO·) production, and reduced retinal vascular leakage. In addition, Western blotting in human retinal microvascular endothelial cells indicated that gCTRP3 triggered AMP-activated protein kinase-α (AMPKα), hence increasing the endothelial NO synthase (eNOS) level and NO· production. In addition, incubation with gCTRP3 in vitro ameliorated the endothelial dysfunction induced by high glucose in the branch of the mesenteric artery. Blockade of either eNOS or AMPKα completely abolished the effects of gCTRP3 described above. Taken together, we demonstrate for the first time that gCTRP3 improves impaired vasodilatation of microvasculature in diabetes by ameliorating endothelial cell function through the AMPK/eNOS/NO· signaling pathway. This finding may suggest an effective intervention against diabetes-associated microvascular complications.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Adipoquinas/farmacología , Diabetes Mellitus Tipo 2/fisiopatología , Células Endoteliales/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/metabolismo , Transducción de Señal/efectos de los fármacos , Adipoquinas/sangre , Adipoquinas/metabolismo , Animales , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Humanos , Masculino , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/metabolismo , Arterias Mesentéricas/fisiología , Ratones Endogámicos C57BL , Microvasos/citología , Factores de Necrosis Tumoral/metabolismo , Vasodilatación/efectos de los fármacos
10.
Am J Physiol Heart Circ Physiol ; 322(1): H71-H86, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34767485

RESUMEN

Microvessels-on-a-chip have enabled in vitro studies to closely simulate in vivo microvessel environment. However, assessing microvessel permeability, a functional measure of microvascular exchange, has not been attainable in nonpermeable microfluidic platforms. This study developed a new approach that enables permeability coefficients (Ps) to be quantified in microvessels developed in nonpermeable chip platforms by integrating avidin-biotin technology. Microvessels were developed on biotinylated fibronectin-coated microfluidic channels. Solute transport was assessed by perfusing microvessels with fluorescence-labeled avidin. Avidin molecules that crossed endothelium were captured by substrate biotin and recorded with real-time confocal images. The Ps was derived from the rate of avidin-biotin accumulation at the substrate relative to solute concentration difference across microvessel wall. Avidin tracers with different physiochemical properties were used to characterize the barrier properties of the microvessel wall. The measured baseline Ps and inflammatory mediator-induced increases in Ps and endothelial cell (EC) [Ca2+]i resembled those observed in intact microvessels. Importantly, the spatial accumulation of avidin-biotin at substrate defines the transport pathways. Glycocalyx layer is well formed on endothelium and its degradation increased transcellular transport without affecting EC junctions. This study demonstrated that in vitro microvessels developed in this simply designed microfluidics structurally possess in vivo-like glycocalyx layer and EC junctions and functionally recapitulate basal barrier properties and stimuli-induced responses observed in intact microvessels. This new approach overcomes the limitations of nonpermeable microfluidics and provides an easily executed highly reproducible in vitro microvessel model with in vivo microvessel functionality, suitable for a wide range of applications in blood and vascular research and drug development.NEW & NOTEWORTHY Our study developed a novel method that allows permeability coefficient to be measured in microvessels developed in nonpermeable microfluidic platforms using avidin-biotin technology. It overcomes the major limitation of nonpermeable microfluidic system and provides a simply designed easily executed and highly reproducible in vitro microvessel model with permeability accessibility. This model with in vivo-like endothelial junctions, glycocalyx, and permeability properties advances microfluidics in microvascular research, suitable for a wide range of biomedical and clinical applications.


Asunto(s)
Avidina , Biotina , Permeabilidad Capilar , Dispositivos Laboratorio en un Chip , Microfluídica/métodos , Microvasos/metabolismo , Animales , Calcio/metabolismo , Células Cultivadas , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Glicocálix/metabolismo , Microfluídica/instrumentación , Microvasos/citología , Ratas
11.
Neurosci Lett ; 770: 136381, 2022 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-34906568

RESUMEN

BACKGROUND: The maintenance of human brain microvascular endothelial cell (HBMEC) function is crucial to improve the outcomes of ischemic stroke (IS). Emerging evidence shows that circular RNAs (circRNAs) are involved in IS progression. This study aimed to investigate the role of circRNA FUN14 domain containing 1 (circFUNDC1) in oxygen-glucose deprivation (OGD)-treated HBMECs. METHODS: The expression of circFUNDC1, miR-375 and phosphatase and tensin homolog (PTEN) mRNA was detected by quantitative real-time PCR (qPCR). Cell viability, apoptosis, migration and angiogenesis were determined by CCK-8 assay, flow cytometry assay, transwell assay and tube formation assay. The protein level of PTEN was detected by western blot. The relationship between miR-375 and circFUNDC1 or PTEN was confirmed by pull-down assay, dual-luciferase reporter assay and RIP assay. Exosomes were identified by transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA). RESULTS: CircFUNDC1 expression was increased in peripheral blood of IS patients and OGD-treated HBMECs. CircFUNDC1 knockdown alleviated OGD-induced cell apoptosis and promoted OGD-blocked cell viability, migration and angiogenesis of HBMECs. MiR-375 was a target of circFUNDC1, and miR-375 restoration played similar effects with circFUNDC1 knockdown. The inhibition of miR-375 reversed the effects of circFUNDC1 knockdown. In addition, PTEN was a downstream target of miR-375, and PTEN overexpression abolished the effects of miR-375 restoration. The expression of circFUNDC1 was elevated in serum-derived exosomes of IS patients, and circFUNDC1 harbored diagnostic values. CONCLUSION: CircFUNDC1 knockdown alleviates OGD-induced HBMECs injuries by inhibiting PTEN via enriching miR-375.


Asunto(s)
Células Endoteliales/metabolismo , Accidente Cerebrovascular Isquémico/metabolismo , Proteínas de la Membrana/genética , MicroARNs/metabolismo , Proteínas Mitocondriales/genética , Fosfohidrolasa PTEN/genética , ARN Circular/metabolismo , Anciano , Encéfalo/irrigación sanguínea , Hipoxia de la Célula , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Exosomas/metabolismo , Femenino , Glucosa/deficiencia , Humanos , Accidente Cerebrovascular Isquémico/genética , Masculino , MicroARNs/genética , Microvasos/citología , Microvasos/metabolismo , Persona de Mediana Edad , Oxígeno/metabolismo , Fosfohidrolasa PTEN/metabolismo , ARN Circular/genética
12.
J Mater Chem B ; 10(2): 271-281, 2022 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-34897348

RESUMEN

Anti-seizure medicines constitute a common yet important modality to treat epilepsy. However, some of them are associated with serious side effects including hepatotoxicity and hypersensitivity. Furthermore, the blood-brain barrier (BBB) is an insurmountable obstacle for brain drug delivery. Fortunately, the introduction of the nanoparticles for drug delivery is a feasible approach to overcome these obstacles. Encapsulating drugs into nanoparticles and delivering them to specific sites shows great potential for improving the efficiency of drug delivery and reducing systemic toxicity. Several in vivo studies have investigated the effect of nanoparticle size on biodistribution in mice, but very few have investigated its effects on efficient drug delivery while crossing the BBB. Therefore, we designed a methoxy poly(lactide-co-glycolide)-b-poly(ethylene glycol) methyl ether (mPEG-PLGA) nanoparticle delivery system and explored the cell uptake efficiency of nanoparticles with different sizes and their ability to penetrate the BBB while carrying carbamazepine (CBZ). CBZ-loaded nanoparticles could significantly reduce the cytotoxicity of CBZ to L929 cells at high concentrations. Results from the endocytosis experiment involving human cerebral microvessel endothelial cell/D3 showed that the DiR-loaded mPEG5K-PLGA10K nanoparticles possessed the highest cell uptake efficiency. The endocytosis efficiency was 90% at 30 min, which far exceeded that of the other groups. Moreover, similar results were obtained from subsequent experiments where fluorescence images of the isolated organs of the mice were acquired. To summarize, our study demonstrated that drug delivery to the brain using nanocarriers is size dependent. Nanoparticles with the smallest particle size can be internalized more effectively, and easily penetrate the BBB, and accumulate in the brain.


Asunto(s)
Anticonvulsivantes/farmacocinética , Barrera Hematoencefálica/fisiología , Carbamazepina/farmacocinética , Portadores de Fármacos/química , Nanopartículas/química , Animales , Anticonvulsivantes/química , Encéfalo/citología , Carbamazepina/química , Línea Celular , Portadores de Fármacos/metabolismo , Portadores de Fármacos/toxicidad , Endocitosis/fisiología , Femenino , Humanos , Ratones , Microvasos/citología , Nanopartículas/metabolismo , Nanopartículas/toxicidad , Tamaño de la Partícula , Poliésteres/química , Poliésteres/metabolismo , Poliésteres/toxicidad , Polietilenglicoles/química , Polietilenglicoles/metabolismo , Polietilenglicoles/toxicidad
13.
Am J Physiol Cell Physiol ; 322(2): C185-C196, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34878922

RESUMEN

The Notch pathway regulates complex patterning events in many species and is critical for the proper formation and function of the vasculature. Despite this importance, how the various components of the Notch pathway work in concert is still not well understood. For example, NOTCH1 stabilizes homotypic endothelial junctions, but the role of NOTCH1 in heterotypic interactions is not entirely clear. NOTCH3, on the other hand, is essential for heterotypic interactions of pericytes with the endothelium, but how NOTCH3 signaling in pericytes impacts the endothelium remains elusive. Here, we use in vitro vascular models to investigate whether pericyte-induced stabilization of the vasculature requires the cooperation of NOTCH1 and NOTCH3. We observe that both pericyte NOTCH3 and endothelial NOTCH1 are required for the stabilization of the endothelium. Loss of either NOTCH3 or NOTCH1 decreases the accumulation of VE-cadherin at endothelial adherens junctions and increases the frequency of wider, more motile junctions. We found that DLL4 was the key ligand for simulating NOTCH1 activation in endothelial cells and observed that DLL4 expression in pericytes is dependent on NOTCH3. Altogether, these data suggest that an interplay between pericyte NOTCH3 and endothelial NOTCH1 is critical for pericyte-induced vascular stabilization.


Asunto(s)
Células Endoteliales/metabolismo , Microvasos/metabolismo , Pericitos/metabolismo , Receptor Notch1/metabolismo , Receptor Notch3/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/farmacología , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al Calcio/farmacología , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/efectos de los fármacos , Células HEK293 , Humanos , Microvasos/citología , Microvasos/efectos de los fármacos , Pericitos/efectos de los fármacos , Receptor Notch1/agonistas , Receptor Notch3/agonistas
14.
Front Immunol ; 12: 759176, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34956190

RESUMEN

Pulmonary microvascular endothelial cells (PMECs) and the extracellular vesicles (EVs) derived from PMECs participate in maintaining pulmonary homeostasis and mediating the inflammatory response. However, obtaining a high-purity population of PMECs and their EVs from mouse is still notoriously difficult. Herein we provide a method to isolate primary mouse PMECs (pMPMECs) and to transduce SV40 lentivirus into pMPMECs to establish an immortalized cell line (iMPMECs), which provides sufficient quantities of EVs for further studies. pMPMECs and iMPMECs can be identified using morphologic criteria, a phenotypic expression profile (e.g., CD31, CD144, G. simplicifolia lectin binding), and functional properties (e.g., Dil-acetylated low-density protein uptake, Matrigel angiogenesis). Furthermore, pMPMEC-EVs and iMPMEC-EVs can be identified and compared. The characteristics of pMPMEC-EVs and iMPMEC-EVs are ascertained by transmission electron microscopy, nanoparticle tracking analysis, and specific protein markers. iMPMECs produce far more EVs than pMPMECs, while their particle size distribution is similar. Our detailed protocol to isolate and immortalize MPMECs will provide researchers with an in vitro model to investigate the specific roles of EVs in pulmonary physiology and diseases.


Asunto(s)
Células Endoteliales/química , Vesículas Extracelulares/química , Microvasos/química , Animales , Células Cultivadas , Células Endoteliales/citología , Células Endoteliales/inmunología , Vesículas Extracelulares/inmunología , Ratones , Microvasos/citología , Microvasos/inmunología , Tamaño de la Partícula , Análisis de la Célula Individual
15.
Dev Cell ; 56(22): 3042-3051, 2021 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-34813766

RESUMEN

Adult organs are vascularized by specialized blood vessels. In addition to inter-organ vascular heterogeneity, each organ is arborized by structurally and functionally diversified populations of endothelial cells (ECs). The molecular pathways that are induced to orchestrate inter- and intra- organ vascular heterogeneity and zonation are shaped during development and fully specified postnatally. Notably, intra-organ specialization of ECs is associated with induction of angiocrine factors that guide cross-talk between ECs and parenchymal cells, establishing co-zonated vascular regions within each organ. In this review, we describe how microenvironmental tissue-specific biophysical, biochemical, immune, and inflammatory cues dictate the specialization of ECs with zonated functions. We delineate how physiological and biophysical stressors in the developing liver, lung, and kidney vasculature induce specialization of capillary beds. Deciphering mechanisms by which vascular microvasculature diversity is attained could set the stage for treating regenerative disorders and promote healing of organs without provoking fibrosis.


Asunto(s)
Células Endoteliales/citología , Riñón/irrigación sanguínea , Microvasos/citología , Neovascularización Fisiológica/fisiología , Regeneración/fisiología , Animales , Diferenciación Celular/fisiología , Humanos , Riñón/citología
16.
Int J Mol Sci ; 22(20)2021 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-34681708

RESUMEN

Ischemia-reperfusion injury (IRI) is an inevitable consequence of organ transplant procedure and associated with acute and chronic organ rejection in transplantation. IRI leads to various forms of programmed cell death, which worsens tissue damage and accelerates transplant rejection. We recently demonstrated that necroptosis participates in murine cardiac microvascular endothelial cell (MVEC) death and murine cardiac transplant rejection. However, MVEC death under a more complex IRI model has not been studied. In this study, we found that simulating IRI conditions in vitro by hypoxia, reoxygenation and treatment with inflammatory cytokines induced necroptosis in MVECs. Interestingly, the apoptosis-inducing factor (AIF) translocated to the nucleus during MVEC necroptosis, which is regulated by the mitochondrial permeability molecule cyclophilin D (CypD). Furthermore, CypD deficiency in donor cardiac grafts inhibited AIF translocation and mitigated graft IRI and rejection (n = 7; p = 0.002). Our studies indicate that CypD and AIF play significant roles in MVEC necroptosis and cardiac transplant rejection following IRI. Targeting CypD and its downstream AIF may be a plausible approach to inhibit IRI-caused cardiac damage and improve transplant survival.


Asunto(s)
Factor Inductor de la Apoptosis/metabolismo , Necroptosis , Peptidil-Prolil Isomerasa F/metabolismo , Animales , Factor Inductor de la Apoptosis/antagonistas & inhibidores , Factor Inductor de la Apoptosis/genética , Hipoxia de la Célula , Núcleo Celular/metabolismo , Peptidil-Prolil Isomerasa F/deficiencia , Peptidil-Prolil Isomerasa F/genética , Células Endoteliales/citología , Células Endoteliales/metabolismo , Interferón gamma/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Microvasos/citología , Modelos Biológicos , Necroptosis/efectos de los fármacos , Oxígeno/farmacología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Factor de Necrosis Tumoral alfa/farmacología
17.
Int J Mol Sci ; 22(19)2021 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-34638868

RESUMEN

Mechanical unloading contributes to significant cardiovascular deconditioning. Endothelial dysfunction in the sites of microcirculation may be one of the causes of the cardiovascular degeneration induced by unloading, but the detailed mechanism is still unclear. Here, we first demonstrated that mechanical unloading inhibited brain microvascular endothelial cell proliferation and downregulated histone deacetylase 6 (HDAC6) expression. Furthermore, HDAC6 promoted microvascular endothelial cell proliferation and attenuated the inhibition of proliferation caused by clinorotation unloading. To comprehensively identify microRNAs (miRNAs) that are regulated by HDAC6, we analyzed differential miRNA expression in microvascular endothelial cells after transfection with HDAC6 siRNA and selected miR-155-5p, which was the miRNA with the most significantly increased expression. The ectopic expression of miR-155-5p inhibited microvascular endothelial cell proliferation and directly downregulated Ras homolog enriched in brain (RHEB) expression. Moreover, RHEB expression was downregulated under mechanical unloading and was essential for the miR-155-5p-mediated promotion of microvascular endothelial cell proliferation. Taken together, these results are the first to elucidate the role of HDAC6 in unloading-induced cell growth inhibition through the miR-155-5p/RHEB axis, suggesting that the HDAC6/miR-155-5p/RHEB pathway is a specific target for the preventative treatment of cardiovascular deconditioning.


Asunto(s)
Proliferación Celular , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Histona Desacetilasa 6/metabolismo , MicroARNs/biosíntesis , Microvasos/metabolismo , Proteína Homóloga de Ras Enriquecida en el Cerebro/metabolismo , Animales , Línea Celular , Células Endoteliales/citología , Ratones , Microvasos/citología
18.
PLoS One ; 16(9): e0256734, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34469488

RESUMEN

OBJECTIVES: This study is to investigate whether the cardiac microvascular endothelial cells (CMECs) can regulate the autophagy of cardiomyocytes (CMs) by secreting lncRNA-ANRIL/miR-181b exosomes, thus participating in the occurrence of uremic cardiovascular disease (CVD). METHODS: A 5/6 nephrectomy uremia model was established, with the mice injected with ANRIL-shRNA lentivirus vector, miR-181b agomir, and related control reagents, containing the serum creatinine and urea nitrogen measured. The renal tissue sections of mice were stained with Periodic Acid-Schiff (PAS), TUNEL, and Hematoxylin-Eosin (HE) performed on myocardial tissue sections of mice. ANRIL-shRNA, miR-181b mimics, and related control reagents were transfected into CMECs, in which the exosomes were extracted and co-cultured with CMs. The expressions of ANRIL, miR-181b and ATG5 were detected by qRT-PCR, and the expressions of autophagy related proteins by Western blot, as well as the binding of ANRIL and miR-181b by the double luciferase reporter gene experiment. RESULTS: ANRIL down-regulation or miR-181b up-regulation can increase the weight of mice with uremia, as well as the expressions of p62 and miR-181b, and reduce the content of serum creatinine and urea nitrogen, the damage of kidney and myocardial tissues, the number of apoptotic cells in myocardial tissues, as well as the expressions of ANRIL, ATG5, Beclin1, and LC3. CMs can absorb the exosomes of CMECs. Compared with IS+ CMEC-Exo group, the expressions of ANRIL and ATG5 in CMs of IS+ CMEC-Exo + sh lncRNA ANRIL and IS+CMEC-Exo+miR-181b mimics groups was down-regulated, as well as the expressions of ATG5, Beclin1, and LC3, while miR-181b expression was up-regulated as well as P62 expression. CONCLUSIONS: CMECs can regulate autophagy of CMs by releasing exosomes containing ANRIL and miR-181b.


Asunto(s)
Proteína 5 Relacionada con la Autofagia/genética , Autofagia/genética , MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Uremia/inmunología , Animales , Proteína 5 Relacionada con la Autofagia/metabolismo , Vasos Coronarios/citología , Vasos Coronarios/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo/inmunología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Exosomas/metabolismo , Humanos , Masculino , Ratones , MicroARNs/genética , Microvasos/citología , Miocardio/citología , Miocardio/inmunología , Miocardio/patología , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/patología , ARN Largo no Codificante/genética , Regulación hacia Arriba/inmunología , Uremia/genética , Uremia/patología
19.
Nat Biomed Eng ; 5(8): 847-863, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34385693

RESUMEN

The therapeutic efficacy of stem cells transplanted into an ischaemic brain depends primarily on the responses of the neurovascular unit. Here, we report the development and applicability of a functional neurovascular unit on a microfluidic chip as a microphysiological model of ischaemic stroke that recapitulates the function of the blood-brain barrier as well as interactions between therapeutic stem cells and host cells (human brain microvascular endothelial cells, pericytes, astrocytes, microglia and neurons). We used the model to track the infiltration of a number of candidate stem cells and to characterize the expression levels of genes associated with post-stroke pathologies. We observed that each type of stem cell showed unique neurorestorative effects, primarily by supporting endogenous recovery rather than through direct cell replacement, and that the recovery of synaptic activities is correlated with the recovery of the structural and functional integrity of the neurovascular unit rather than with the regeneration of neurons.


Asunto(s)
Accidente Cerebrovascular Isquémico/terapia , Dispositivos Laboratorio en un Chip , Trasplante de Células Madre , Astrocitos/citología , Astrocitos/metabolismo , Barrera Hematoencefálica/química , Barrera Hematoencefálica/metabolismo , Técnicas de Cocultivo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Humanos , Microglía/citología , Microglía/metabolismo , Microvasos/citología , Modelos Biológicos , Neuronas/citología , Neuronas/metabolismo , Pericitos/citología , Pericitos/metabolismo , Células Madre/citología , Células Madre/metabolismo
20.
Molecules ; 26(16)2021 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-34443317

RESUMEN

Endothelial cell injury is an early event in systemic sclerosis (SSc) pathogenesis and several studies indicate oxidative stress as the trigger of SSc-associated vasculopathy. Here, we show that circulating factors present in sera of SSc patients increased reactive oxygen species (ROS) production and collagen synthesis in human pulmonary microvascular endothelial cells (HPMECs). In addition, the possibility that iloprost, a drug commonly used in SSc therapy, might modulate the above-mentioned biological phenomena has been also investigated. In this regard, as compared to sera of SSc patients, sera of iloprost-treated SSc patients failed to increased ROS levels and collagen synthesis in HPMEC, suggesting a potential antioxidant mechanism of this drug.


Asunto(s)
Colágeno/biosíntesis , Células Endoteliales/efectos de los fármacos , Iloprost/farmacología , Microvasos/citología , Estrés Oxidativo/efectos de los fármacos , Esclerodermia Sistémica/sangre , Suero/metabolismo , Adulto , Células Endoteliales/citología , Células Endoteliales/metabolismo , Femenino , Humanos , Masculino , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...