Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
Discov Med ; 35(176): 264-274, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37272093

RESUMO

BACKGROUND: Amonafide (Amo), due to hematotoxicity and digestive tract symptoms, the clinical application of which is limited. Several studies have reported that chemotherapy side effects are closely related to cellular senescence accumulation. Our study aims to examine whether amonafide causes senescence in human umbilical vein endothelial cell (HUVEC) lines and investigate its mechanisms associated with senescence. METHODS: The experiments of expression of genes and proteins associated with aging were carried out with HUVEC cell lines. The experiments were divided into a control group and an amonafide group with different days. The HUVEC senescence cells were detected by SA-ß-Gal staining, Western blotting detected the protein levels of p16, p53, AMPK (Adenosine 5'-Monophosphate (AMP)-Activated Protein Kinase), mTOR (mechanistic Target of Rapamycin), p62, and LC3 (microtubule-associated protein1 light chain 3, MAP1LC3). Fluorescence detected the expression of mRFP (monomeric Red Fluorescent Protein)-GFP (Green Fluorescent Protein)-LC3 and LC3 puncta of HUVEC cells. RT-qPCR (Real-Time Quantitative Polymerase Chain Reaction) tested the expressions of p53, p21, IL (Interleukin)-1ß, IL-6 (Interleukin-6), IL-8 (Interleukin-8), and MCP-1 (Monocyte Chemoattractant Protein-1). CCK-8 (Cell Counting Kit-8) assessed the HUVEC cell viability. RESULTS: Here, we reported that amonafide resulted in an increased proportion of SA-ß-Gal positive cells, high expression of aging-related proteins (p53 p < 0.05; p16 p < 0.05), and aging-related genes (p53 p < 0.05; p21 p < 0.05; IL-1ß p < 0.05; IL-6 p < 0.05; IL-8 p < 0.05; MCP-1 p < 0.05) on the 3rd day. Mechanistically, amonafide could cause an increase in the levels of the mTOR (p < 0.05) on days 1 and 3, and p62 protein (p < 0.05) on day 1, and a decline in LC3II (microtubule-associated protein1 light chain 3Ⅱ)/LC3I levels (p < 0.05) on day 3, which is associated with the regulation of senescence. Additionally, the viability of HUVECs (human umbilical vein endothelial cells) was significantly inhibited by amonafide starting with a concentration of 0.8 µm (p < 0.05). CONCLUSIONS: We first discovered that amonafide caused normal cellular senescence in our experiments. Amonafide-induced cellular aging by inhibiting autophagy and activating the mTOR pathway. The findings may offer new strategies for managing adverse reactions to amonafide.


Assuntos
Interleucina-8 , Proteína Supressora de Tumor p53 , Humanos , Células Endoteliais da Veia Umbilical Humana/química , Células Endoteliais da Veia Umbilical Humana/metabolismo , Interleucina-8/metabolismo , Interleucina-8/farmacologia , Proteína Supressora de Tumor p53/análise , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Interleucina-6/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Autofagia , Senescência Celular/fisiologia
2.
ACS Appl Mater Interfaces ; 14(1): 439-451, 2022 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-34942074

RESUMO

Additive manufacturing is a promising technology for the fabrication of customized implants with complex geometry. The objective of this study was to investigate the initial cell-material interaction of degradable Fe-30Mn-1C-0.02S stent structures in comparison to conventional 316L as a reference, both processed by laser powder bed fusion. FeMn-based alloys have comparable mechanical properties with clinically applied AISI 316L for a corrosion-resistant stent material. Different corrosion stages of the as-built Fe-30Mn-1C-0.02S stent surfaces were simulated by pre-conditioning in DMEM under cell culture conditions for 2 h, 7 days, and 28 days. Human umbilical vein endothelial cells (HUVECs) were directly seeded onto the pre-conditioned samples, and cell viability, adherence, and morphology were analyzed. These studies were accompanied by measurements of iron and manganese ion release and Auger electron spectroscopy to evaluate the influence of corrosion products and degradation on the cells. In the initial phase (2 h of pre-conditioning), HUVECs were able to attach but the cell number decreased over the cultivation period of 14 days and the CD31 staining pattern of intercellular contacts was disordered. At later time points of corrosion (7 and 28 days of pre-conditioning), CD31 staining was distinctly located at the intercellular contacts, and the cell density increased after seeding and was stable for up to 14 days. Formation of a complex degradation layer, which had a composition and thickness dependent on the pre-conditioning time, led to a reduced ion release and finally showed a positive effect on cell survival. Concluding, our data suggest the suitability of Fe-30Mn-1C-0.02S for in vivo applications.


Assuntos
Materiais Biocompatíveis/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Ferro/metabolismo , Lasers , Manganês/metabolismo , Materiais Biocompatíveis/química , Células Cultivadas , Células Endoteliais da Veia Umbilical Humana/química , Humanos , Íons/química , Íons/metabolismo , Ferro/química , Manganês/química , Teste de Materiais
3.
Eur Cytokine Netw ; 33(4): 79-89, 2022 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37227141

RESUMO

BACKGROUND AND OBJECTIVE: Endothelial cell activation plays a critical role in leukocyte recruitment during inflammation and infection. We previously found that cholinergic stimulation (via vagus nerve stimulation) attenuates vascular endothelial impairment and reduces the inflammatory profile in ovariectomized rats. However, the specific molecular mechanism is unclear. This study was designed to explore the effects and molecular mechanisms of cholinergic agonists (acetylcholine [ACh]) on lipopolysaccharide (LPS)-induced endothelial cell activation in vitro. METHODS: Human umbilical vein endothelial cells (HUVECs) were treated with different concentrations of LPS (10/100/1000 ng/mL) to activate endothelial cells. HUVECs were untreated, treated with ACh (10-5 M) alone, treated with 100 ng/mL LPS alone, or treated with different concentrations of ACh (10-9/10-8/10-7/10-6/10-5 M) before LPS stimulation. HUVECs were also pre-treated with 10-6 M ACh with or without mecamylamine (an nAChR blocker) (10 µΜ) and methyllycaconitine (a specific α7 nAChR blocker) (10 µΜ) and incubated with or without LPS. ELISA, western blotting, cell immunofluorescence, and cell adhesion assays were used to examine inflammatory cytokine production, adhesion molecule expression, monocyte-endothelial cell adhesion and activation of the MAPK/NF-κB pathways. RESULTS: LPS (at 10 ng/mL, 100 ng/mL and 1,000 ng/mL) increased VCAM-1 expression in HUVECs in a dose-dependent manner (with no significant difference between LPS at 100 ng/mL and 1,000 ng/mL). ACh (10-9 M-10-5 M) blocked adhesion molecule expression (VCAM-1, ICAM-1, and E-selectin) and inflammatory cytokine production (TNF-α, IL-6, MCP-1, IL-8) in response to LPS in a dose-dependent manner (with no significant difference between 10-5 and 10-6 M Ach). LPS was also shown to significantly enhance monocyte-endothelial cell adhesion, which was largely abrogated by treatment with ACh (10-6M). VCAM-1 expression was blocked by mecamylamine rather than methyllycaconitine. Lastly, ACh (10-6 M) significantly reduced LPS-induced phosphorylation of NF-κB/p65, IκBα, ERK, JNK and p38 MAPK in HUVECs, which was blocked by mecamylamine. CONCLUSIONS: ACh protects against LPS-induced endothelial cell activation by inhibiting the MAPK and NF-κB pathways, which are mediated by nAChR, rather than α7 nAChR. Our results may provide novel insight into the anti-inflammatory effects and mechanisms of ACh.


Assuntos
Lipopolissacarídeos , NF-kappa B , Animais , Humanos , Ratos , Acetilcolina/farmacologia , Acetilcolina/metabolismo , Células Endoteliais da Veia Umbilical Humana/química , Células Endoteliais da Veia Umbilical Humana/metabolismo , Molécula 1 de Adesão Intercelular/análise , Molécula 1 de Adesão Intercelular/metabolismo , Lipopolissacarídeos/farmacologia , Mecamilamina/metabolismo , Mecamilamina/farmacologia , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo , Molécula 1 de Adesão de Célula Vascular/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos
4.
J Cell Mol Med ; 25(15): 7436-7450, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34235869

RESUMO

Exosomes are secreted into the extracellular space by most cell types and contain various molecular constituents, which play roles in many biological processes. Adipose-derived mesenchymal stem cells (ADSCs) can differentiate into a variety of cell types and secrete a series of paracrine factors through exosomes. ADSC-derived exosomes have shown diagnostic and therapeutic potential in many clinical diseases. The molecular components are critical for their mechanisms. Several methods have been developed for exosome purification, including ultracentrifugation, ultrafiltration, density gradient purification, size-based isolation, polymer precipitation and immuno-affinity purification. Thus, we employed four methods to isolate exosomes from the hADSC culture medium, including ultracentrifugation, size exclusion chromatography, ExoQuick-TC precipitation and ExoQuick-TC ULTRA isolation. Following exosome isolation, we performed quantitative proteomic analysis of the exosome proteins using isobaric tags for relative and absolute quantification (iTRAQ) labelling, combined with 2D-LC-MS/MS. There were 599 universal and 138 stably expressed proteins in hADSC-derived exosomes. We proved that these proteins were potential hADSC-derived exosomes markers, including CD109, CD166, HSPA4, TRAP1, RAB2A, RAB11B and RAB14. From the quantitative proteomic analysis, we demonstrated that hADSC-derived exosome protein expression varied, with lipopolysaccharide (LPS) treatment, in the different isolation methods. Pathway analysis and proliferation, migration and endothelial tube formation assays showed varying effects in cells stimulated with hADSC-derived exosomes from different isolation methods. Our study revealed that different isolation methods might introduce variations in the protein composition in exosomes, which reflects their effects on biological function. The pros and cons of these methods are important points to consider for downstream research applications.


Assuntos
Fracionamento Celular/métodos , Exossomos/química , Células-Tronco Mesenquimais/química , Proteoma/química , Proteômica/métodos , Adipócitos/química , Células Cultivadas , Exossomos/metabolismo , Células Endoteliais da Veia Umbilical Humana/química , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Redes e Vias Metabólicas
5.
ACS Appl Bio Mater ; 4(5): 4084-4093, 2021 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-35006825

RESUMO

A critical component of tissue engineering is the ability to functionally replace native tissue stroma. Electrospinning is a technique capable of forming fibrous constructs with a high surface area for increased cell-material interaction and enhanced biocompatibility. However, physical and biological properties of electrospun scaffolds are limited by design controllability on a macroscale. We developed a methodology for generating electrospun scaffolds with defined patterns and topographic features to influence physical properties and biological interactions. Five unique design electrospinning target collectors were fabricated to allow for generation of defined polymeric scaffold patterns including lines, sinusoids, squares, zigzags, and solid. Poly(lactic-co-glycolic) acid was electrospun under identical conditions utilizing these varied targets, and constructs generated were examined as to their physical configuration, mechanical and chemical properties, and their ability to foster vascular smooth muscle cell adhesion and retention at 24 h. Modifying collector designs led to significant differences in fiber target coverage ranging from 300 mm2 for solid (100% of the target area) to 217.8 mm2 for lines (72.6% of the target area). Measured fiber excess, residual open area, and contact angle (hydrophobicity) followed the same trend as fiber target coverage with respect to the collector pattern: lines > sinusoids > squares > zigzags > solid. Similarly, the line design allowed for the greatest cell adhesion and retention (258 ± 31 cells), whereas solid exhibited the lowest (150 ± 15 cells); p < 0.05. There was a strong direct correlation of cell adhesion to construct residual open area (R2 = 0.94), normalized fiber excess (R2 = 0.99), and fiber grammage (R2 = 0.72), with an inverse relationship to fiber target coverage (R2 = 0.94). Our results demonstrate the ability to utilize patterned collectors for modifying macroscopic and microscopic electrospun scaffold features, which directly impact cell adhesion and retention, offering translational utility for designing specific tissue constructs.


Assuntos
Materiais Biocompatíveis/química , Células Endoteliais da Veia Umbilical Humana/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Adesão Celular , Células Cultivadas , Humanos , Teste de Materiais , Tamanho da Partícula
6.
Am J Physiol Cell Physiol ; 320(2): C216-C224, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33326314

RESUMO

Leukocyte adhesion to the endothelium is an important early step in the initiation and progression of sepsis. The endothelial glycocalyx layer (EGL) has been implicated in neutrophil adhesion and barrier dysfunction, but studies in this area are few. In this report, we examine the hypothesis that damage to the structure of the EGL caused by inflammation leads to increased leukocyte adhesion and endothelial barrier dysfunction. We used human umbilical vein endothelial cells enzymatically treated to remove the EGL components hyaluronic acid (HA) and heparan sulfate (HS) as a model for EGL damage. Using atomic force microscopy, we show reductions in EGL thickness after removal of either HA or HS individually, but the largest decrease, comparable with TNF-α treatment, was observed when both HA and HS were removed. Interestingly, removal of HS or HA individually did not affect neutrophil adhesion significantly, but removal of both constituents resulted in increased neutrophil adhesion. To test EGL contributions to endothelial barrier properties, we measured transendothelial electrical resistance (TEER) and diffusion of fluorescently labeled dextran (10 kDa molecular weight) across the monolayer. Removal of EGL components decreased TEER but had an insignificant effect on dextran diffusion rates. The reduction in TEER suggests that disruption of the EGL may predispose endothelial cells to increased rates of fluid leakage. These data support the view that damage to the EGL during inflammation has significant effects on the accessibility of adhesion molecules, likely facilitates leukocyte adhesion, and may also contribute to increased rates of fluid transport into tissues.


Assuntos
Citoproteção/fisiologia , Glicocálix/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Mediadores da Inflamação/metabolismo , Mediadores da Inflamação/toxicidade , Citoproteção/efeitos dos fármacos , Glicocálix/química , Glicocálix/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/química , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Neutrófilos/química , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo
7.
ACS Appl Mater Interfaces ; 12(40): 44563-44577, 2020 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-32966746

RESUMO

Three-dimensional (3D) bioprinting of hydrogel-based constructs at adequate consistency and reproducibility can be obtained through a compromise between the hydrogel's inherent instability and printing fidelity. There is an increasing demand to develop bioprinting modalities that enable high-fidelity fabrication of 3D hydrogel structures that closely correspond to the envisioned design. In this work, we performed a systematic, in-depth characterization and optimization of embedded 3D bioprinting to create 3D gelatin-methacryloyl (gelMA) structures with highly controlled fidelity using Carbopol as suspension bath. The role of various embedded printing process parameters in bioprinting fidelity was investigated using a combination of experimental and theoretical approaches. We examined the effect of rheological properties of gelMA and Carbopol at varying concentrations, as well as printing conditions on the volumetric flow rate of gelMA bioink. Printing speed was examined and optimized to successfully print gelMA into the support bath at varying Carbopol concentrations. Printing fidelity was characterized in terms of printed strand diameter, uniformity, angle, and area. The optimal Carbopol solution that retained filament shape at highest fidelity was determined. The efficacy of developed bioprinting approach was then demonstrated by fabricating 3D hydrogel constructs with varying geometries and visualized using an advanced synchrotron-based imaging technique. We also investigated the influence of the Carbopol medium on cross-linking and the resulting stiffness of gelMA constructs. Finally, in vitro cytotoxicity of the developed bioprinting approach was assessed by printing human umbilical vein endothelial cells encapsulated in the gelMA bioink. These results demonstrate the significance of the close interplay between bioink-support bath rheology and printing parameters and help to establish an optimized workflow for creating 3D hydrogel structures with high fidelity and cytocompatibility via embedded bioprinting techniques. This robust platform could further expand the application of bioprinted soft tissue constructs in a wide variety of biomedical applications.


Assuntos
Resinas Acrílicas/análise , Metacrilatos/química , Impressão Tridimensional , Células Cultivadas , Gelatina/química , Células Endoteliais da Veia Umbilical Humana/química , Humanos , Tamanho da Partícula , Propriedades de Superfície
8.
Anal Biochem ; 600: 113760, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32353372

RESUMO

Hydrogen peroxide is an unavoidable by-product of cell metabolism, but when it is not properly managed by the body it can lead to several pathologies (e.g., premature aging, cardiovascular and neurodegenerative diseases, cancer). Several methods have been proposed for the measurement of intracellular H2O2 but none of them has proven to be selective. We developed a rapid all-in-one chemiluminescent bioassay for the quantification of H2O2 in living cells with a low limit of detection (0.15 µM). The method relies on an adamantylidene-1,2-dioxetane lipophilic probe containing an arylboronate moiety; upon reaction with H2O2 the arylboronate moiety is converted to the correspondent phenol and the molecule decomposes leading to an excited-state fragment that emits light. The probe has been successfully employed for quantifying intracellular H2O2 in living human endothelial, colon and keratinocyte cells exposed to different pro-oxidant stimuli (i.e., menadione, phorbol myristate acetate and lipopolysaccharide). Imaging experiments clearly localize the chemiluminescence emission inside the cells. Treatment of cells with antioxidant molecules leads to a dose-dependent decrease of intracellular H2O2 levels. As a proof of concept, the bioassay has been used to measure the antioxidant activity of extracts from Brassica juncea wastes, which contain glucosinolates, isothiocyanates and other antioxidant molecules.


Assuntos
Corantes Fluorescentes/química , Células Endoteliais da Veia Umbilical Humana/química , Peróxido de Hidrogênio/análise , Medições Luminescentes , Imagem Óptica , Células CACO-2 , Células Cultivadas , Humanos , Estrutura Molecular
9.
J Colloid Interface Sci ; 571: 326-336, 2020 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-32208203

RESUMO

Templated assembly techniques have been extensively used to develop various types of hemoglobin (Hb) loaded particles with improved performance. However, several instability issues must still be solved, including Hb exposure, enhanced Hb auto-oxidation, and the relatively weak binding of Hb to cross-linkers. Herein, to meet the stability requirements for novel hemoglobin-based oxygen carriers (HBOCs), hemoglobin-polydopamine particles (Hb-PDA) were fabricated using a mild process that combines the co-precipitation of Hb and an inorganic template with the spontaneous adhesion of PDA. The Hb-PDA showed uniform size distribution, chemical integrity of both Hb and PDA, high biocompatibility, and robust oxygen delivery. Our results demonstrated that the use of polydopamine as a biocompatible coating material reduced Hb leakage from the particles under both static and flow conditions, thus mitigating the toxicity associated with free Hb and strengthening the stability of Hb particles. In addition, Hb-PDA reduced HUVEC (Human Umbilical Vein Cells) oxidative injury and scavenged 85% of the available hydroxyl radicals, exhibiting its potential to act as an antioxidant for encapsulated Hb. Hb-PDA therefore shows significant promise as a cell-like structurally and functionally stable HBOCs.


Assuntos
Materiais Revestidos Biocompatíveis/metabolismo , Hemoglobinas/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Indóis/metabolismo , Oxigênio/metabolismo , Polímeros/metabolismo , Materiais Revestidos Biocompatíveis/química , Hemoglobinas/química , Células Endoteliais da Veia Umbilical Humana/química , Humanos , Indóis/química , Oxigênio/química , Tamanho da Partícula , Polímeros/química , Propriedades de Superfície
10.
Chem Commun (Camb) ; 56(18): 2731-2734, 2020 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-32022000

RESUMO

We herein develop two ß-galactosidase (ß-Gal) activatable NIR fluorescent probes for visualizing ovarian cancers. Particularly, probe BOD-M-ßGal produced NIR-II emission light at 900-1300 nm upon ß-Gal activation. By using our activatable and target specific NIR-II probe for deep-tissue imaging of ß-Gal overexpressed ovarian cancer cells, rapid and accurate imaging of ovarian tumors in nude mice was achieved.


Assuntos
Corantes Fluorescentes/química , Imagem Óptica , Neoplasias Ovarianas/diagnóstico por imagem , beta-Galactosidase/química , Animais , Linhagem Celular Tumoral , Ativação Enzimática , Feminino , Corantes Fluorescentes/metabolismo , Células Endoteliais da Veia Umbilical Humana/química , Humanos , Raios Infravermelhos , Camundongos , Camundongos Nus , Estrutura Molecular , Neoplasias Experimentais/diagnóstico por imagem , Neoplasias Experimentais/metabolismo , Neoplasias Ovarianas/metabolismo , beta-Galactosidase/metabolismo
11.
Int Wound J ; 17(1): 65-72, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31665826

RESUMO

Platelets contain abundant growth factors and cytokines that have a positive influence on the migration and proliferation of different cell types by modulating its physiopathological processes. As it is known that human umbilical cord blood platelet lysate (UCB-PL) contains a supraphysiological concentration of growth factors, in the present study, we investigated its effectiveness in wound-healing processes. Human UCB-PL was obtained by the freeze/thaw of platelet concentrate (1.1 × 109 platelets/L), and its effect was evaluated on human or mouse endothelial cells, monocytes, fibroblasts, and keratinocytes in different concentrations. Human UCB-PL was observed to have high levels of pro-angiogenic growth factor than peripheral blood platelet-rich plasma. Among the cell lines, different concentrations of human UCB-PL were necessary to influence their viability and proliferation. For L929 cells, 5% of total volume was necessary, while for human umbilical vein endothelial cell, it was 10%. Cell migration on monocytes was increased with respect to the positive control, and scratch closure on keratinocytes was increased with respect to serum-free medium with only 10% of human UCB-PL. We concluded that the human UCB-PL may be useful to produce a large amount of standard platelet concentrates sufficient for several clinical-scale expansions avoiding inter-individual variability, which can also be used as a functional tool for clinical regenerative application for wound healing.


Assuntos
Plaquetas/química , Proliferação de Células/efeitos dos fármacos , Citocinas/uso terapêutico , Fatores de Crescimento Endotelial/uso terapêutico , Células Endoteliais da Veia Umbilical Humana/química , Plasma Rico em Plaquetas/química , Cicatrização/efeitos dos fármacos , Ferimentos e Lesões/tratamento farmacológico , Proliferação de Células/fisiologia , Células Cultivadas/efeitos dos fármacos , Humanos , Cicatrização/fisiologia
12.
Apoptosis ; 25(1-2): 73-91, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31768842

RESUMO

Weightlessness-induced cardiovascular dysfunction can lead to physiological and pathological consequences. It has been shown that spaceflight or simulated microgravity can alter expression profiles of some microRNAs (miRNAs). Here, we attempt to identify the role of miRNAs in human umbilical vein endothelial cells (HUVECs) apoptosis under simulated microgravity. RNA-sequencing and quantitative real-time PCR (qRT-PCR) assays were used to identify differentially expressed miRNAs in HUVECs under simulated microgravity. Then we obtained the target genes of these miRNAs through target analysis software. Moreover, GO and KEGG enrichment analysis were performed. The effects of these miRNAs on HUVECs apoptosis were evaluated by flow cytometry, Western blot and Hoechst staining. Furthermore, we obtained the target gene of miR-27b-5p by luciferase assay, qRT-PCR and Western blot. Finally, we investigated the relationship between this target gene and miR-27b-5p in HUVECs apoptosis under normal gravity or simulated microgravity. We found 29 differentially expressed miRNAs in HUVECs under simulated microgravity. Of them, the expressions of 3 miRNAs were validated by qRT-PCR. We demonstrated that miR-27b-5p affected HUVECs apoptosis by inhibiting zinc fingers and homeoboxes 1 (ZHX1). Our results reported here demonstrate for the first time that simulated microgravity can alter the expression of some miRNAs in HUVECs and miR-27b-5p may protect HUVECs from apoptosis under simulated microgravity by targeting ZHX1.


Assuntos
Apoptose , Células Endoteliais da Veia Umbilical Humana/citologia , Ausência de Peso/efeitos adversos , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Células Endoteliais da Veia Umbilical Humana/química , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
13.
Biofabrication ; 12(1): 015004, 2019 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-31470437

RESUMO

The ability to fabricate perfusable, small-diameter vasculature is a foundational step toward generating human tissues/organs for clinical applications. Currently, it is highly challenging to generate vasculature integrated with smooth muscle and endothelium that replicates the complexity and functionality of natural vessels. Here, a novel method for directly printing self-standing, small-diameter vasculature with smooth muscle and endothelium is presented through combining tailored mussel-inspired bioink and unique 'fugitive-migration' tactics, and its effectiveness and advantages over other methods (i.e. traditional alginate/calcium hydrogel, post-perfusion of endothelial cells) are demonstrated. The biologically inspired, catechol-functionalized, gelatin methacrylate (GelMA/C) undergoes rapid oxidative crosslinking in situ to form an elastic hydrogel, which can be engineered with controllable mechanical strength, high cell/tissue adhesion, and excellent bio-functionalization. The results demonstrate the bioprinted vascular construct possessed numerous favorable, biomimetic characteristics such as proper biomechanics, higher tissue affinity, vascularized tissue manufacturing ability, beneficial perfusability and permeability, excellent vasculoactivity, and in vivo autonomous connection (∼2 weeks) as well as vascular remodeling (∼6 weeks). The advanced achievements in creating biomimetic, functional vasculature illustrate significant potential toward generating a complicated vascularized tissue/organ for clinical transplantation.


Assuntos
Bioimpressão/métodos , Células Endoteliais da Veia Umbilical Humana/citologia , Músculo Liso/citologia , Alginatos/química , Bioimpressão/instrumentação , Gelatina/química , Células Endoteliais da Veia Umbilical Humana/química , Humanos , Hidrogéis/química , Músculo Liso/química , Impressão Tridimensional , Engenharia Tecidual , Alicerces Teciduais/química
14.
Anal Chem ; 91(17): 11023-11029, 2019 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-31266308

RESUMO

Förster resonance energy transfer (FRET) by using fluorescent carbon dots (CDs) as energy donors shows potential for biosensing and bioimaging. However, it remains underused and underestimated for CDs as a building block for FRET owing to the low efficiency and complex operation originating from the surface modification of CDs. To overcome these limitations, herein we develop a novel FRET soft nanoball (fretSNB) in which thousands of green CDs and black hole quencher 2 (BHQ-2) dyes are loaded, and FRET occurs from CDs to BHQ-2 dyes with the consequence of effective fluorescence quenching. These fretSNBs can be ruptured in the presence of phospholipase A2 (PLA2) released in a process of duplex-specific nuclease (DSN)-assisted target recycling amplification (TRA), making the fluorescence of CDs recovered. Thus, a dual amplification strategy is successfully developed for amplified detection of microribonucleic acids (miRNAs) in the concentration range 0.025-10 nM with a limit of detection (3σ) reaching 16.5 pM which is about 515 times lower than without fretSNBs. In addition, the developed strategy exhibits high selectivity for discrimination of a single nucleotide difference and capability to detect miRNAs extracted from cells, suggesting excellent potential in biomedical analysis and clinical diagnosis.


Assuntos
Técnicas Biossensoriais , Transferência Ressonante de Energia de Fluorescência/métodos , Nanopartículas Metálicas/química , MicroRNAs/análise , Técnicas de Amplificação de Ácido Nucleico/métodos , Compostos Azo/química , Linhagem Celular , Células Epiteliais/química , Células Epiteliais/metabolismo , Células HeLa , Células Endoteliais da Veia Umbilical Humana/química , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Limite de Detecção , MicroRNAs/genética , MicroRNAs/metabolismo , Fosfolipases A2/química , Pontos Quânticos
15.
Sci Rep ; 9(1): 7203, 2019 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-31076589

RESUMO

Endothelial cells have multifaceted interactions with the immune system, both as initiators and targets of immune responses. In vivo, apoptotic endothelial cells release two types of extracellular vesicles upon caspase-3 activation: apoptotic bodies and exosome-like nanovesicles (ApoExos). Only ApoExos are immunogenic: their injection causes inflammation and autoimmunity in mice. Based on deep sequencing of total RNA, we report that apoptotic bodies and ApoExos are loaded with divergent RNA cargos that are not released by healthy endothelial cells. Apoptotic bodies, like endothelial cells, contain mainly ribosomal RNA whereas ApoExos essentially contain non-ribosomal non-coding RNAs. Endogenous retroelements, bearing viral-like features, represented half of total ApoExos RNA content. ApoExos also contained several copies of unedited Alu repeats and large amounts of non-coding RNAs with a demonstrated role in autoimmunity such as U1 RNA and Y RNA. Moreover, ApoExos RNAs had a unique nucleotide composition and secondary structure characterized by strong enrichment in U-rich motifs and unstably folded RNAs. Globally, ApoExos were therefore loaded with RNAs that can stimulate a variety of RIG-I-like receptors and endosomal TLRs. Hence, apoptotic endothelial cells selectively sort in ApoExos a diversified repertoire of immunostimulatory "self RNAs" that are tailor-made for initiation of innate immune responses and autoimmunity.


Assuntos
Vesículas Extracelulares/genética , Perfilação da Expressão Gênica/métodos , Células Endoteliais da Veia Umbilical Humana/citologia , RNA/imunologia , Apoptose , Proteína DEAD-box 58/metabolismo , Células Endoteliais da Veia Umbilical Humana/química , Humanos , RNA/genética , Edição de RNA , Receptores Imunológicos , Análise de Sequência de RNA , Receptores Toll-Like/metabolismo
16.
Mol Med Rep ; 19(3): 1603-1612, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30628687

RESUMO

Endothelial dysfunction is a key pathophysiological step in early stage diabetes mellitus (DM) macrovascular complications and is also crucial in the inflammatory mechanisms of macrovascular complications. However, there is currently no effective intervention to improve endothelial dysfunction associated with DM macrovascular complications. Astragaloside IV (AS­IV), which can be extracted from the traditional Chinese medicine Astragalus membranaceus, has potential therapeutic effects on DM and its complications. The present study evaluated the effect of AS­IV on high glucose­induced human umbilical vein endothelial cell (HUVEC) injury and its possible mechanism. The result indicated that AS­IV has a significant protective effect on high glucose­induced HUVEC injury. AS­IV could significantly promote cell proliferation, reduce apoptosis and decrease the protein and mRNA expression levels of tumor necrosis factor­α and interleukin­1ß in HUVECs. Furthermore, AS­IV could decrease the expression of phosphorylated c­Jun NH2­terminal kinase (JNK) phosphorylated apoptosis signal­regulating kinase 1, cytochrome c, cleaved­caspase­9, cleaved­caspase­3 and the relative ratio of B­cell lymphoma­2 associated X protein/B­cell lymphoma­2 in HUVECs. In conclusion, the present study demonstrated that AS­IV could suppress apoptosis and inflammatory reactions promoted by high glucose conditions in HUVECs by inhibiting the JNK signaling pathway. These findings suggest that AS­IV could inhibit the process of endothelial dysfunction in diabetic macrovascular complications.


Assuntos
Astragalus propinquus/química , Inflamação/tratamento farmacológico , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Saponinas/farmacologia , Triterpenos/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Glucose/toxicidade , Células Endoteliais da Veia Umbilical Humana/química , Humanos , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/patologia , Saponinas/química , Transdução de Sinais/efeitos dos fármacos , Triterpenos/química
17.
J Pept Sci ; 25(5): e3146, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30652389

RESUMO

The placental growth factor (PlGF), a member of VEGF family, plays a crucial role in pathological angiogenesis, especially ischemia, inflammation, and cancer. This activity is mediated by its selective binding to VEGF receptor 1 (VEGFR-1), which occurs predominantly through receptor domains 2 and 3. The PlGF ß-hairpin region spanning residues Q87 to V100 is one of the key binding elements on the protein side. We have undertaken a study on the design, preparation, and functional characterization of the peptide reproducing this region and of a set of analogues where glycine 94, occurring at the corner of the hairpin in the native protein, is replaced by charged as well as hydrophobic residues. Also, some peptides with arginine 96 replaced by other residues have been studied. We find that the parent peptide weakly binds VEGFR-1, but replacement of G94 with residues bearing H-bond donating residues significantly improves the affinity. Replacement of R96 instead blocks the interaction between the peptide and the domain. The strongest affinity is observed with the G94H (peptide PlGF-2) and G94W (peptide PlGF-10) mutants, while the peptide PlGF-8, bearing the R96G mutation, is totally inactive. The PlGF-1 and PlGF-2 peptides also bind the VEGFR-2 receptors, though with a reduced affinity, and are able to interfere with the VEGF-induced receptor signaling on endothelial cells. The peptides also bind VEGFR-2 on the surface of cells, while PlGF-8 is inactive. Data suggest that these peptides have potential applications as PlGF/VEGF mimic in various experimental settings.


Assuntos
Células Endoteliais da Veia Umbilical Humana/química , Proteínas de Membrana/química , Peptídeos/química , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/química , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/química , Sítios de Ligação , Proliferação de Células , Células Endoteliais , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Peptídeos/metabolismo , Propriedades de Superfície , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
18.
J Colloid Interface Sci ; 539: 351-360, 2019 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-30594010

RESUMO

Salvianic acid-loaded mesoporous silica nanoparticles into gelatin/polyurethane bilayered small-diameter tubular scaffold were prepared by thermally induced phase separation (TIPS) and electrospinning. Mesoporous silica nanoparticles (MSNs) were selected as carriers to load salvianic acid (SAL). The SAL-loaded MSNs (SAL@MSNs) with an optimized SAL loading efficiency of 10% was initially dispersed in gelatin solution and under a vacuum freeze-drying process as an inner layer of vascular scaffolds. Then, poly(ester-urethane)urea (C-PEEUU) nanofibers were electrospun outside the SAL@MSNs/Gelatin vascular scaffold to strengthen the spongy matrix. The loaded SAL within the MSNs/Gelatin/C-PEEUU bilayered small-diameter tubular scaffold showed a sustained release profile and good mechanical properties. In addition, the drug-loaded composite scaffold showed no unfavorable effects on the adhesion and proliferation of endothelial cells. Moreover, no intimal hyperplasia and acute thrombosis was observed in the short-term implantation in rabbit's carotid artery. We believe the SAL@MSNs/Gelatin/C-PEEUU bilayered vascular scaffolds have promise for vascular tissue engineering applications.


Assuntos
Implante de Prótese Vascular , Artérias Carótidas/química , Células Endoteliais da Veia Umbilical Humana/química , Engenharia Tecidual , Alicerces Teciduais , Alcenos/química , Animais , Artérias Carótidas/citologia , Adesão Celular , Proliferação de Células , Células Cultivadas , Feminino , Gelatina/química , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Nanopartículas/química , Tamanho da Partícula , Polifenóis/química , Poliuretanos/química , Porosidade , Coelhos , Dióxido de Silício/química , Propriedades de Superfície
19.
J Agric Food Chem ; 66(48): 12866-12875, 2018 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-30406651

RESUMO

Neovascularization causes serious oculopathy related to upregulation of vascular-endothelial-growth factor (VEGF) causing new capillary growth via endothelial cells. Green-tea-extract (GTE) constituents possess antiangiogenesis properties. We used VEGF to induce human umbilical-vein endothelial cells (HUVECs) and applied GTE, epigallocatechin gallate (EGCG), and mixtures of different compositions of purified catechins (M1 and M2) to evaluate their efficacies of inhibition and their underlying mechanisms using cell-cycle analysis and untargeted metabolomics techniques. GTE, EGCG, M1, and M2 induced HUVEC apoptosis by 22.1 ± 2, 20.0 ± 0.7, 50.7 ± 8.5, and 69.8 ± 4.1%, respectively. GTE exerted a broad, balanced metabolomics spectrum, involving suppression of the biosynthesis of cellular building blocks and oxidative-phosphorylation metabolites as well as promotion of the biosynthesis of membrane lipids and growth factors. M2 mainly induced mechanisms associated with energy and biosynthesis suppression. Therefore, GTE exerted mechanisms involving both promotion and suppression activities, whereas purified catechins induced extensive apoptosis. GTE could be a more promising antineovascularization remedy for ocular treatment.


Assuntos
Camellia sinensis/química , Catequina/análogos & derivados , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Extratos Vegetais/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Apoptose/efeitos dos fármacos , Catequina/farmacologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/química , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Lipídeos de Membrana/biossíntese , Metabolômica
20.
Analyst ; 143(17): 4180-4188, 2018 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-30070265

RESUMO

Although the plasma membrane is a major site for nitric oxide (NO) generation and action, few targetable probes that specifically sense and image NO in the plasma membrane have been reported. In this study, a membrane targetable, two-photon nitric oxide probe, Mem-NO, was developed and evaluated for bio-imaging of both exogenous and endogenous NO. By installing a quaternary ammonium compound as the hydrophilic head and a long alkyl chain as the hydrophobic tail on 4-amino-1,8-naphthalimide, we designed Mem-NO into a bi-polar structure. Due to the interaction with the phospholipid bilayer of plasma membrane, Mem-NO could specifically and stably localize in the plasma membrane. Mem-NO is almost nonfluorescent, but it displayed substantial fluorescence enhancement (16-fold) upon NO capture with sensitive (74 nM limit of detection) and fast response (within 1 min). Moreover, Mem-NO displayed strong two-photon excitation fluorescence activity (δ = 177 GM at 810 nm) and low cytotoxicity. It was found that Mem-NO is capable of two-photon imaging of endogenous NO in live neurons and human umbilical vein endothelial cells (HUVECs) and exogenous NO in mouse brain tissues. Therefore, Mem-NO qualifies as an essential and unique analytical tool for monitoring NO for future physiological, pathological, and pharmacological studies.


Assuntos
Química Encefálica , Membrana Celular/química , Corantes Fluorescentes , Óxido Nítrico/análise , Animais , Células Endoteliais da Veia Umbilical Humana/química , Humanos , Camundongos , Neurônios/química , Imagem Óptica , Células PC12 , Fótons , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA