Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 6.999
Filtrer
1.
J Biomed Mater Res B Appl Biomater ; 112(7): e35448, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38968133

RÉSUMÉ

Traditional decellularized bioscaffolds possessing intact vascular networks and unique architecture have been extensively studied as conduits for repairing nerve damage. However, they are limited by the absence of electrical conductivity, which is crucial for proper functioning of nervous tissue. This study focuses on investigating decellularized umbilical cord arteries by applying coatings of graphene oxide (GO) and reduced graphene oxide (RGO) to their inner surfaces. This resulted in a homogeneous GO coating that fully covered the internal lumen of the arteries. The results of electrical measurements demonstrated that the conductivity of the scaffolds could be significantly enhanced by incorporating RGO and GO conductive sheets. At a low frequency of 0.1 Hz, the electrical resistance level of the coated scaffolds decreased by 99.8% with RGO and 98.21% with GO, compared with uncoated scaffolds. Additionally, the mechanical properties of the arteries improved by 24.69% with GO and 32.9% with RGO after the decellularization process. The GO and RGO coatings did not compromise the adhesion of endothelial cells and promoted cell growth. The cytotoxicity tests revealed that cell survival rate increased over time with RGO, while it decreased with GO, indicating the time-dependent effect on the cytotoxicity of GO and RGO. Blood compatibility evaluations showed that graphene nanomaterials did not induce hemolysis but exhibited some tendency toward blood coagulation.


Sujet(s)
Matériaux revêtus, biocompatibles , Conductivité électrique , Graphite , Artères ombilicales , Graphite/composition chimique , Humains , Matériaux revêtus, biocompatibles/composition chimique , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Structures d'échafaudage tissulaires/composition chimique , Test de matériaux , Cordon ombilical/cytologie , Animaux
2.
Clin Lab ; 70(7)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38965970

RÉSUMÉ

BACKGROUND: In this study, we aimed to identify the hub genes responsible for increased vascular endothelial cell permeability. METHODS: We applied the weighted Gene Expression Omnibus (GEO) database to mine dataset GSE178331 and ob-tained the most relevant high-throughput sequenced genes for an increased permeability of vascular endothelial cells due to inflammation. We constructed two weighted gene co-expression network analysis (WGCNA) networks, and the differential expression of high-throughput sequenced genes related to endothelial cell permeability were screened from the GEO database. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were performed on the differential genes. Their degree values were obtained from the topological properties of protein-protein interaction (PPI) networks of differential genes, and the hub genes associated with an increased endothelial cell permeability were analyzed. Reverse transcription-polymerase chain reaction (RT-PCR) and western blotting techniques were used to detect the presence of these hub genes in TNF-α induced mRNA and the protein expression in endothelial cells. RESULTS: In total, 1,475 differential genes were mainly enriched in the cell adhesion and TNF-α signaling pathway. With TNF-α inducing an increase in the endothelial cell permeability and significantly increasing mRNA and protein expression levels, we identified three hub genes, namely PTGS2, ICAM1, and SNAI1. There was a significant difference in the high-dose TNF-α group and in the low-dose TNF-α group compared to the control group, in the endothelial cell permeability experiment (p = 0.008 vs. p = 0.02). Measurement of mRNA and protein levels of PTGS2, ICAM1, and SNAI1 by western blotting analysis showed that there was a significant impact on TNF-α and that there was a significant dose-dependent relationship (p < 0.05 vs. p < 0.01). CONCLUSIONS: The three hub genes identified through bioinformatics analyses in the present study may serve as biomarkers of increased vascular endothelial cell permeability. The findings offer valuable insights into the progress and mechanism of vascular endothelial cell permeability.


Sujet(s)
Biologie informatique , Cellules endothéliales , Réseaux de régulation génique , Cartes d'interactions protéiques , Facteur de nécrose tumorale alpha , Humains , Biologie informatique/méthodes , Facteur de nécrose tumorale alpha/génétique , Facteur de nécrose tumorale alpha/métabolisme , Cellules endothéliales/métabolisme , Analyse de profil d'expression de gènes/méthodes , Cyclooxygenase 2/génétique , Cyclooxygenase 2/métabolisme , Perméabilité capillaire , Transduction du signal , Bases de données génétiques , Molécule-1 d'adhérence intercellulaire/génétique , Molécule-1 d'adhérence intercellulaire/métabolisme , Facteurs de transcription de la famille Snail/génétique , Facteurs de transcription de la famille Snail/métabolisme , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Gene Ontology
3.
J Cell Mol Med ; 28(10): e18402, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-39008328

RÉSUMÉ

Syntaxin 17 (STX17) has been identified as a crucial factor in mediating the fusion of autophagosomes and lysosomes. However, its specific involvement in the context of atherosclerosis (AS) remains unclear. This study sought to elucidate the role and mechanistic contributions of STX17 in the initiation and progression of AS. Utilizing both in vivo and in vitro AS model systems, we employed ApoE knockout (KO) mice subjected to a high-fat diet and human umbilical vein endothelial cells (HUVECs) treated with oxidized low-density lipoprotein (ox-LDL) to assess STX17 expression. To investigate underlying mechanisms, we employed shRNA-STX17 lentivirus to knock down STX17 expression, followed by evaluating autophagy and inflammation in HUVECs. In both in vivo and in vitro AS models, STX17 expression was significantly upregulated. Knockdown of STX17 exacerbated HUVEC damage, both with and without ox-LDL treatment. Additionally, we observed that STX17 knockdown impaired autophagosome degradation, impeded autophagy flux and also resulted in the accumulation of dysfunctional lysosomes in HUVECs. Moreover, STX17 knockdown intensified the inflammatory response following ox-LDL treatment in HUVECs. Further mechanistic exploration revealed an association between STX17 and STING; reducing STX17 expression increased STING levels. Further knockdown of STING enhanced autophagy flux. In summary, our findings suggest that STX17 knockdown worsens AS by impeding autophagy flux and amplifying the inflammatory response. Additionally, the interaction between STX17 and STING may play a crucial role in STX17-mediated autophagy.


Sujet(s)
Athérosclérose , Autophagie , Cellules endothéliales de la veine ombilicale humaine , Inflammation , Lipoprotéines LDL , Protéines Qa-SNARE , Autophagie/génétique , Animaux , Humains , Athérosclérose/métabolisme , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Inflammation/métabolisme , Inflammation/anatomopathologie , Inflammation/génétique , Protéines Qa-SNARE/métabolisme , Protéines Qa-SNARE/génétique , Souris , Lipoprotéines LDL/métabolisme , Techniques de knock-down de gènes , Lysosomes/métabolisme , Souris knockout , Mâle , Souris de lignée C57BL , Modèles animaux de maladie humaine , Alimentation riche en graisse/effets indésirables , Apolipoprotéines E/génétique , Apolipoprotéines E/métabolisme , Apolipoprotéines E/déficit
4.
Scand J Immunol ; 99(4): e13354, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-39008522

RÉSUMÉ

Systemic sclerosis (SSc) is a chronic autoimmune connective tissue disease. Vascular damage is one of the important features of SSc, which affects the progression and prognosis of the disease. MiR-126-3p is an important microRNA (miRNA) that regulates vascular structure and function, which can be transported through exosomes. However, the role of miR-126-3p in vascular damage in SSc is still unclear. Therefore, we focused on the connection between miR-126-3p and vascular damage in SSc, as well as investigated the potential role of miR-126-3p in vascular damage in SSc. First, this study successfully extracted extracellular vesicles from clinical plasma samples and characterized the exosomes within them. Then, we predicted and screened the target pathway mammalian/mechanistic target of rapamycin (mTOR) and the target gene SLC7A5 of miR-126-3p through online databases. Next, we constructed SSc mice for in vivo studies. The results showed that the expression of miR-126-3p was decreased in the plasma exosomes, while the SLC7A5 expression, autophagy, and lipid peroxidation were increased in the aorta. Luciferase reporter gene assays demonstrated that miR-126-3p can bind to SLC7A5, resulting in a decrease in its expression. In vitro experiments have shown that exosomal miR-126-3p can be internalized by human umbilical vein endothelial cells (HUVECs). The miR-126-3p group exhibited enhanced cell viability and tube formation ability, along with increased expression of the vascular formation marker CD31. Additionally, miR-126-3p downregulated the protein expression of SLC7A5 and LC3 in HUVECs, while upregulating the protein expression of mTOR, P62, PPARγ, and CPT-1. However, the effects of miR-126-3p on HUVECs were counteracted by mTOR inhibitors and enhanced by mTOR activators. The results indicated that exosomal miR-126-3p has the potential to protect against vascular injury in SSc by regulating the SLC7A5/mTOR signalling pathway in HUVECs.


Sujet(s)
Exosomes , Cellules endothéliales de la veine ombilicale humaine , microARN , Transduction du signal , Sérine-thréonine kinases TOR , microARN/génétique , microARN/métabolisme , Humains , Exosomes/métabolisme , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Animaux , Souris , Sclérodermie systémique/métabolisme , Sclérodermie systémique/génétique , Sclérodermie systémique/anatomopathologie , Mâle , Femelle , Transporteur-1 d'acides aminés neutres à longue chaîne/métabolisme , Transporteur-1 d'acides aminés neutres à longue chaîne/génétique , Adulte d'âge moyen , Modèles animaux de maladie humaine , Adulte
5.
J Extracell Vesicles ; 13(7): e12449, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39001708

RÉSUMÉ

Hypoxia is a common feature of solid tumours and activates adaptation mechanisms in cancer cells that induce therapy resistance and has profound effects on cellular metabolism. As such, hypoxia is an important contributor to cancer progression and is associated with a poor prognosis. Metabolic alterations in cells within the tumour microenvironment support tumour growth via, amongst others, the suppression of immune reactions and the induction of angiogenesis. Recently, extracellular vesicles (EV) have emerged as important mediators of intercellular communication in support of cancer progression. Previously, we demonstrated the pro-angiogenic properties of hypoxic cancer cell derived EV. In this study, we investigate how (hypoxic) cancer cell derived EV mediate their effects. We demonstrate that cancer derived EV regulate cellular metabolism and protein synthesis in acceptor cells through increased activation of mTOR and AMPKα. Using metabolic tracer experiments, we demonstrate that EV stimulate glucose uptake in endothelial cells to fuel amino acid synthesis and stimulate amino acid uptake to increase protein synthesis. Despite alterations in cargo, we show that the effect of cancer derived EV on recipient cells is primarily determined by the EV producing cancer cell type rather than its oxygenation status.


Sujet(s)
AMP-Activated Protein Kinases , Vésicules extracellulaires , Glycolyse , Tumeurs , Biosynthèse des protéines , Sérine-thréonine kinases TOR , Humains , Sérine-thréonine kinases TOR/métabolisme , AMP-Activated Protein Kinases/métabolisme , Vésicules extracellulaires/métabolisme , Tumeurs/métabolisme , Cellules endothéliales/métabolisme , Glucose/métabolisme , Lignée cellulaire tumorale , Microenvironnement tumoral , Cellules endothéliales de la veine ombilicale humaine/métabolisme
6.
Sci Rep ; 14(1): 16270, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39009650

RÉSUMÉ

Steroid-induced osteonecrosis of the femoral head (SONFH) is the predominant cause of non-traumatic osteonecrosis of the femoral head (ONFH). Impaired blood supply and reduced osteogenic activity of the femoral head are the key pathogenic mechanisms of SONFH. Fibroblast growth factor 23 (FGF23) levels are not only a biomarker for early vascular lesions caused by abnormal mineral metabolism, but can also act directly on the peripheral vascular system, leading to vascular pathology. The aim of this study was to observe the role of FGF23 on bone microarchitecture and vascular endothelium, and to investigate activation of pyroptosis in SONFH. Lipopolysaccharide (LPS) combined with methylprednisolone (MPS) was applied for SONFH mouse models, and adenovirus was used to increase or decrease the level of FGF23. Micro-CT and histopathological staining were used to observe the structure of the femoral head, and immunohistochemical staining was used to observe the vascular density. The cells were further cultured in vitro and placed in a hypoxic environment for 12 h to simulate the microenvironment of vascular injury during SONFH. The effect of FGF23 on osteogenic differentiation was evaluated using alkaline phosphatase staining, alizarin red S staining and expression of bone formation-related proteins. Matrigel tube formation assay in vitro and immunofluorescence were used to detect the ability of FGF23 to affect endothelial cell angiogenesis. Steroids activated the pyroptosis signaling pathway, promoted the secretion of inflammatory factors in SONFH models, led to vascular endothelial dysfunction and damaged the femoral head structure. In addition, FGF23 inhibited the HUVECs angiogenesis and BMSCs osteogenic differentiation. FGF23 silencing attenuated steroid-induced osteonecrosis of the femoral head by inhibiting the pyroptosis signaling pathway, and promoting osteogenic differentiation of BMSCs and angiogenesis of HUVECs in vitro.


Sujet(s)
Nécrose de la tête fémorale , Facteur-23 de croissance des fibroblastes , Facteurs de croissance fibroblastique , Ostéogenèse , Pyroptose , Pyroptose/effets des médicaments et des substances chimiques , Facteur-23 de croissance des fibroblastes/métabolisme , Animaux , Nécrose de la tête fémorale/induit chimiquement , Nécrose de la tête fémorale/métabolisme , Nécrose de la tête fémorale/anatomopathologie , Souris , Facteurs de croissance fibroblastique/métabolisme , Ostéogenèse/effets des médicaments et des substances chimiques , Humains , Tête du fémur/anatomopathologie , Tête du fémur/métabolisme , Modèles animaux de maladie humaine , Méthylprednisolone/pharmacologie , Mâle , Lipopolysaccharides/toxicité , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Différenciation cellulaire , Stéroïdes/pharmacologie
7.
J Am Heart Assoc ; 13(14): e034076, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-38958135

RÉSUMÉ

BACKGROUND: Endothelial cell (EC) dysfunction involves reduced nitric oxide (NO) bioavailability due to NO synthase uncoupling linked to increased oxidation and reduced cofactor availability. Loss of endothelial function and NO bioavailability are associated with inflammation, including leukocyte activation. Eicosapentaenoic acid (EPA) administered as icosapent ethyl reduced cardiovascular events in REDUCE-IT (Reduction of Cardiovascular Events With Icosapent Ethyl-Intervention Trial) in relation to on-treatment EPA blood levels. The mechanisms of cardiovascular protection for EPA remain incompletely elucidated but likely involve direct effects on the endothelium. METHODS AND RESULTS: In this study, human ECs were treated with EPA and challenged with the cytokine IL-6 (interleukin-6). Proinflammatory responses in the ECs were confirmed by ELISA capture of sICAM-1 (soluble intercellular adhesion molecule-1) and TNF-α (tumor necrosis factor-α). Global protein expression was determined using liquid chromatography-mass spectrometry tandem mass tag. Release kinetics of NO and peroxynitrite were monitored using porphyrinic nanosensors. IL-6 challenge induced proinflammatory responses from the ECs as evidenced by increased release of sICAM-1 and TNF-α, which correlated with a loss of NO bioavailability. ECs pretreated with EPA modulated expression of 327 proteins by >1-fold (P<0.05), compared with IL-6 alone. EPA augmented expression of proteins involved in NO production, including heme oxygenase-1 and dimethylarginine dimethylaminohydrolase-1, and 34 proteins annotated as associated with neutrophil degranulation. EPA reversed the endothelial NO synthase uncoupling induced by IL-6 as evidenced by an increased [NO]/[peroxynitrite] release ratio (P<0.05). CONCLUSIONS: These direct actions of EPA on EC functions during inflammation may contribute to its distinct cardiovascular benefits.


Sujet(s)
Acide eicosapentanoïque , Inflammation , Interleukine-6 , Monoxyde d'azote , Facteur de nécrose tumorale alpha , Humains , Acide eicosapentanoïque/analogues et dérivés , Acide eicosapentanoïque/pharmacologie , Monoxyde d'azote/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Interleukine-6/métabolisme , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Molécule-1 d'adhérence intercellulaire/métabolisme , Heme oxygenase-1/métabolisme , Nitric oxide synthase type III/métabolisme , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Cellules cultivées , Biodisponibilité , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Acide peroxynitreux/métabolisme , Médiateurs de l'inflammation/métabolisme
8.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-39000232

RÉSUMÉ

Various human diseases are triggered by molecular alterations influencing the fine-tuned expression and activity of transcription factors, usually due to imbalances in targets including protein-coding genes and non-coding RNAs, such as microRNAs (miRNAs). The transcription factor EB (TFEB) modulates human cellular networks, overseeing lysosomal biogenesis and function, plasma-membrane trafficking, autophagic flux, and cell cycle progression. In endothelial cells (ECs), TFEB is essential for the maintenance of endothelial integrity and function, ensuring vascular health. However, the comprehensive regulatory network orchestrated by TFEB remains poorly understood. Here, we provide novel mechanistic insights into how TFEB regulates the transcriptional landscape in primary human umbilical vein ECs (HUVECs), using an integrated approach combining high-throughput experimental data with dedicated bioinformatics analysis. By analyzing HUVECs ectopically expressing TFEB using ChIP-seq and examining both polyadenylated mRNA and small RNA sequencing data from TFEB-silenced HUVECs, we have developed a bioinformatics pipeline mapping the different gene regulatory interactions driven by TFEB. We show that TFEB directly regulates multiple miRNAs, which in turn post-transcriptionally modulate a broad network of target genes, significantly expanding the repertoire of gene programs influenced by this transcription factor. These insights may have significant implications for vascular biology and the development of novel therapeutics for vascular disease.


Sujet(s)
Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines , Biologie informatique , Réseaux de régulation génique , Cellules endothéliales de la veine ombilicale humaine , microARN , Humains , microARN/génétique , microARN/métabolisme , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/métabolisme , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/génétique , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Biologie informatique/méthodes , Régulation de l'expression des gènes , Cellules endothéliales/métabolisme
9.
Int J Mol Sci ; 25(13)2024 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-39000349

RÉSUMÉ

Lipid emulsions are used as adjuvant drugs to alleviate intractable cardiovascular collapse induced by drug toxicity. We aimed to examine the effect of lipid emulsions on labetalol-induced vasodilation and the underlying mechanism in the isolated rat aorta. We studied the effects of endothelial denudation, NW-nitro-l-arginine methyl ester (l-NAME), calmidazolium, methylene blue, 1H-[1,2,4]oxadiazolo[4,3-a] quinoxalin-1-one (ODQ), and lipid emulsions on labetalol-induced vasodilation. We also evaluated the effects of lipid emulsions on cyclic guanosine monophosphate (cGMP) formation, endothelial nitric oxide synthase (eNOS) phosphorylation, and endothelial calcium levels induced by labetalol. Labetalol-induced vasodilation was higher in endothelium-intact aortas than that in endothelium-denuded aortas. l-NAME, calmidazolium, methylene blue, and ODQ inhibited labetalol-induced vasodilation in endothelium-intact aortas. Lipid emulsions inhibited labetalol-induced vasodilation in endothelium-intact and endothelium-denuded aortas. l-NAME, ODQ, and lipid emulsions inhibited labetalol-induced cGMP formation in endothelium-intact aortas. Lipid emulsions reversed the stimulatory and inhibitory eNOS (Ser1177 and Thr495) phosphorylation induced by labetalol in human umbilical vein endothelial cells and inhibited the labetalol-induced endothelial calcium increase. Moreover, it decreased labetalol concentration. These results suggest that lipid emulsions inhibit vasodilation induced by toxic doses of labetalol, which is mediated by the inhibition of endothelial nitric oxide release and reduction of labetalol concentration.


Sujet(s)
Aorte , GMP cyclique , Émulsions , Labétalol , Nitric oxide synthase type III , Vasodilatation , Animaux , Vasodilatation/effets des médicaments et des substances chimiques , Rats , Aorte/effets des médicaments et des substances chimiques , Aorte/métabolisme , Labétalol/pharmacologie , Mâle , Nitric oxide synthase type III/métabolisme , GMP cyclique/métabolisme , Endothélium vasculaire/effets des médicaments et des substances chimiques , Endothélium vasculaire/métabolisme , Rat Sprague-Dawley , Humains , Lipides , Phosphorylation/effets des médicaments et des substances chimiques , Calcium/métabolisme , L-NAME/pharmacologie , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/métabolisme
10.
Int J Mol Sci ; 25(13)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-39000362

RÉSUMÉ

Exposure to microgravity during spaceflight induces the alterations in endothelial cell function associated with post-flight cardiovascular deconditioning. PIEZO1 is a major mechanosensitive ion channel that regulates endothelial cell function. In this study, we used a two-dimensional clinostat to investigate the expression of PIEZO1 and its regulatory mechanism on human umbilical vein endothelial cells (HUVECs) under simulated microgravity. Utilizing quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot analysis, we observed that PIEZO1 expression was significantly increased in response to simulated microgravity. Moreover, we found microgravity promoted endothelial cells migration by increasing expression of PIEZO1. Proteomics analysis highlighted the importance of C-X-C chemokine receptor type 4(CXCR4) as a main target molecule of PIEZO1 in HUVECs. CXCR4 protein level was increased with simulated microgravity and decreased with PIEZO1 knock down. The mechanistic study showed that PIEZO1 enhances CXCR4 expression via Ca2+ influx. In addition, CXCR4 could promote endothelial cell migration under simulated microgravity. Taken together, these results suggest that the upregulation of PIEZO1 in response to simulated microgravity regulates endothelial cell migration due to enhancing CXCR4 expression via Ca2+ influx.


Sujet(s)
Mouvement cellulaire , Cellules endothéliales de la veine ombilicale humaine , Canaux ioniques , Récepteurs CXCR4 , Simulation d'apesanteur , Récepteurs CXCR4/métabolisme , Récepteurs CXCR4/génétique , Humains , Canaux ioniques/métabolisme , Canaux ioniques/génétique , Mouvement cellulaire/génétique , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Calcium/métabolisme , Cellules endothéliales/métabolisme , Régulation de l'expression des gènes
11.
Nutrients ; 16(13)2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38999793

RÉSUMÉ

The epigenetic regulation of nuclear factor erythroid 2-related factor 2 (Nrf2), a pivotal redox transcription factor, plays a crucial role in maintaining cellular homeostasis. Recent research has underscored the significance of epigenetic modifications of Nrf2 in the pathogenesis of diabetic foot ulcers (DFUs). This study investigates the epigenetic reversal of Nrf2 by pterostilbene (PTS) in human endothelial cells in a hyperglycemic microenvironment (HGM). The activation potential of PTS on Nrf2 was evaluated through ARE-Luciferase reporter assays and nuclear translocation studies. Following 72 h of exposure to an HGM, mRNA expression and protein levels of Nrf2 and its downstream targets NAD(P)H quinone oxidoreductase 1 (NQO1), heme-oxygenase 1(HO-1), superoxide dismutase (SOD), and catalase (CAT) exhibited a decrease, which was mitigated in PTS-pretreated endothelial cells. Epigenetic markers, including histone deacetylases (HDACs class I-IV) and DNA methyltransferases (DNMTs 1/3A and 3B), were found to be downregulated under diabetic conditions. Specifically, Nrf2-associated HDACs, including HDAC1, HDAC2, HDAC3, and HDAC4, were upregulated in HGM-induced endothelial cells. This upregulation was reversed in PTS-pretreated cells, except for HDAC2, which exhibited elevated expression in endothelial cells treated with PTS in a hyperglycemic microenvironment. Additionally, PTS was observed to reverse the activity of the methyltransferase enzyme DNMT. Furthermore, CpG islands in the Nrf2 promoter were hypermethylated in cells exposed to an HGM, a phenomenon potentially counteracted by PTS pretreatment, as shown by methyl-sensitive restriction enzyme PCR (MSRE-qPCR) analysis. Collectively, our findings highlight the ability of PTS to epigenetically regulate Nrf2 expression under hyperglycemic conditions, suggesting its therapeutic potential in managing diabetic complications.


Sujet(s)
Antioxydants , Cellules endothéliales , Épigenèse génétique , Hyperglycémie , Facteur-2 apparenté à NF-E2 , Stilbènes , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Humains , Épigenèse génétique/effets des médicaments et des substances chimiques , Stilbènes/pharmacologie , Hyperglycémie/métabolisme , Antioxydants/pharmacologie , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Microenvironnement cellulaire/effets des médicaments et des substances chimiques , Histone deacetylases/métabolisme , Histone deacetylases/génétique , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Extinction de l'expression des gènes , Stress oxydatif/effets des médicaments et des substances chimiques , Méthylation de l'ADN/effets des médicaments et des substances chimiques
12.
J Transl Med ; 22(1): 643, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38982516

RÉSUMÉ

BACKGROUND: Diabetic foot ulcer (DFU) is the most devastating complication of diabetes mellitus (DM) and plays a major role in disability and death in DM patients. NADH: ubiquinone oxidoreductase subunit B5 (NDUFB5) plays an important role in maintaining mitochondrial respiration, but whether it is involved in regulating the progression of advanced glycation end products (AGEs)-mediated DFU is still unclear. METHODS: Firstly, the role of AGEs on cell viability, migration, and mitochondrial respiration in human umbilical vein endothelial cells (HUVECs) was explored in vitro. Next, NDUFB5 expression was detected in human samples and AGEs-treated HUVECs, and NDUFB5's effect on AGEs-induced HUVECs injury and skin wound in diabetic mice was further clarified. In addition, the role of m6A modification mediated by methyltransferase-like 3 (METTL3) in regulating NDUFB5 expression and AGEs-induced HUVECs injury was investigated. RESULTS: NDUFB5 promoted cell viability, migration, and mitochondrial respiration in AGEs-treated HUVECs, whereas mitochondrial fusion promoter M1 facilitated cell viability, migration, and mitochondrial oxiadative respiration in NDUFB5 knockdown HUVECs. Meanwhile, NDUFB5 promotes skin wound healing in diabetic mice. Besides, METTL3-mediated m6A modification and insulin like growth factor 2 mRNA binding protein 2 (IGF2BP2) enhanced NDUFB5 expression in HUVECs. Furthermore, METTL3 promoted cell viability, migration, and mitochondrial respiration in AGEs-treated HUVECs by increasing NDUFB5. CONCLUSION: METTL3-mediated NDUFB5 m6A modification inhibits AGEs-induced cell injury in HUVECs. METTL3 and NDUFB5 might serve as potential targets for DFU therapy in the future.


Sujet(s)
Mouvement cellulaire , Pied diabétique , Cellules endothéliales de la veine ombilicale humaine , Methyltransferases , Mitochondries , Cicatrisation de plaie , Humains , Methyltransferases/métabolisme , Animaux , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Mitochondries/métabolisme , Pied diabétique/anatomopathologie , Pied diabétique/métabolisme , Mâle , Respiration cellulaire , Produits terminaux de glycation avancée/métabolisme , Survie cellulaire , Diabète expérimental/métabolisme , Diabète expérimental/complications , Souris , Souris de lignée C57BL
13.
J Cell Mol Med ; 28(13): e18529, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38984945

RÉSUMÉ

In this in vitro study, for the first time, we evaluate the effects of simvastatin-loaded liposome nanoparticles (SIM-LipoNPs) treatment on fibrosis-induced liver microtissues, as simvastatin (SIM) has shown potential benefits in the non-alcoholic fatty liver disease process. We developed multicellular liver microtissues composed of hepatic stellate cells, hepatoblastoma cells and human umbilical vein endothelial cells. The microtissues were supplemented with a combination of palmitic acid and oleic acid to develop fibrosis models. Subsequently, various groups of microtissues were exposed to SIM and SIM-LipoNPs at doses of 5 and 10 mg/mL. The effectiveness of the treatments was evaluated by analysing cell viability, production of reactive oxygen species (ROS) and nitric oxide (NO), the expression of Kruppel-like factor (KLF) 2, and pro-inflammatory cytokines (interleukin(IL)-1 α, IL-1 ß, IL-6 and tumour necrosis factor-α), and the expression of collagen I. Our results indicated that SIM-LipoNPs application showed promising results. SIM-LipoNPs effectively amplified the SIM-klf2-NO pathway at a lower dosage compatible with a high dosage of free SIM, which also led to reduced oxidative stress by decreasing ROS levels. SIM-LipoNPs administration also resulted in a significant reduction in pro-inflammatory cytokines and Collagen I mRNA levels, as a marker of fibrosis. In conclusion, our study highlights the considerable therapeutic potential of using SIM-LipoNPs to prevent liver fibrosis progress, underscoring the remarkable properties of SIM-LipoNPs in activating the KLF2-NO pathway and anti-oxidative and anti-inflammatory response.


Sujet(s)
Cellules étoilées du foie , Facteurs de transcription Krüppel-like , Liposomes , Cirrhose du foie , Nanoparticules , Espèces réactives de l'oxygène , Simvastatine , Humains , Simvastatine/pharmacologie , Cirrhose du foie/traitement médicamenteux , Cirrhose du foie/anatomopathologie , Cirrhose du foie/métabolisme , Espèces réactives de l'oxygène/métabolisme , Nanoparticules/composition chimique , Facteurs de transcription Krüppel-like/métabolisme , Facteurs de transcription Krüppel-like/génétique , Cellules étoilées du foie/effets des médicaments et des substances chimiques , Cellules étoilées du foie/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques , Cytokines/métabolisme , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Monoxyde d'azote/métabolisme
14.
Bull Exp Biol Med ; 177(1): 115-123, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38963596

RÉSUMÉ

The cardiac perivascular niche is a cellular microenvironment of a blood vessel. The principles of niche regulation are still poorly understood. We studied the effect of TGFß1 on cells forming the cardiac perivascular niche using 3D cell culture (cardiospheres). Cardiospheres contained progenitor (c-Kit), endothelial (CD31), and mural (αSMA) cells, basement membrane proteins (laminin) and extracellular matrix proteins (collagen I, fibronectin). TGFß1 treatment decreased the length of CD31+ microvasculature, VE cadherin protein level, and proportion of NG2+ cells, and increased proportion of αSMA+ cells and transgelin/SM22α protein level. We supposed that this effect is related to the stabilizing function of TGFß1 on vascular cells: decreased endothelial cell proliferation, as shown for HUVEC, and activation of mural cell differentiation.


Sujet(s)
Différenciation cellulaire , Prolifération cellulaire , Facteur de croissance transformant bêta-1 , Facteur de croissance transformant bêta-1/pharmacologie , Facteur de croissance transformant bêta-1/métabolisme , Différenciation cellulaire/effets des médicaments et des substances chimiques , Humains , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Animaux , Protéines des microfilaments/métabolisme , Protéines des microfilaments/génétique , Antigènes CD31/métabolisme , Cadhérines/métabolisme , Laminine/métabolisme , Laminine/pharmacologie , Protéines du muscle/métabolisme , Cellules cultivées , Cellules endothéliales/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/cytologie , Fibronectines/métabolisme , Fibronectines/pharmacologie , Antigènes CD/métabolisme , Myocarde/métabolisme , Myocarde/cytologie , Niche de cellules souches/effets des médicaments et des substances chimiques , Niche de cellules souches/physiologie , Collagène de type I/métabolisme , Sphéroïdes de cellules/effets des médicaments et des substances chimiques , Sphéroïdes de cellules/métabolisme , Sphéroïdes de cellules/cytologie , Techniques de cultures cellulaires tridimensionnelles/méthodes
15.
Sci Rep ; 14(1): 15113, 2024 07 02.
Article de Anglais | MEDLINE | ID: mdl-38956421

RÉSUMÉ

The aims of this study were to determine whether human umbilical cord mesenchymal stem cells (hucMSCs) modified by miRNA-25-3p (miR-25-3p) overexpression could promote venous endothelial cell proliferation and attenuate portal endothelial cell injury. HucMSCs and human umbilical vein endothelial cells (HUVEC) were isolated and cultured from human umbilical cord and characterized. Lentiviral vectors expressing miRNA-25-3p were transfected into hucMSCs and confirmed by PCR. We verified the effect of miR-25-3p-modified hucMSCs on HUVEC by cell co-culture and cell supernatant experiments. Subsequently, exosomes of miR-25-3p-modified hucMSCs were isolated from cell culture supernatants and characterized by WB, NTA and TEM. We verified the effects of miR-25-3p-modified exosomes derived from hucMSCs on HUVEC proliferation, migration, and angiogenesis by in vitro cellular function experiments. Meanwhile, we further examined the downstream target genes and signaling pathways potentially affected by miR-25-3p-modified hucMSC-derived exosomes in HUVEC. Finally, we established a rat portal vein venous thrombosis model by injecting CM-DiR-labeled hucMSCs intravenously into rats and examining the homing of cells in the portal vein by fluorescence microscopy. Histological and immunohistochemical experiments were used to examine the effects of miRNA-25-3p-modified hucMSCs on the proliferation and damage of portal vein endothelial cells. Primary hucMSCs and HUVECs were successfully isolated, cultured and characterized. Primary hucMSCs were modified with a lentiviral vector carrying miR-25-3p at MOI 80. Co-culture and cell supernatant intervention experiments showed that overexpression of miRNA-25-3p in hucMSCs enhanced HUVEC proliferation, migration and tube formation in vitro. We successfully isolated and characterized exosomes of miR-25-3p-modified hucMSCs, and exosome intervention experiments demonstrated that miR-25-3p-modified exosomes derived from hucMSCs similarly enhanced the proliferation, migration, and angiogenesis of HUVECs. Subsequent PCR and WB analyses indicated PTEN/KLF4/AKT/ERK1/2 as potential pathways of action. Analysis in a rat portal vein thrombosis model showed that miR-25-3p-modified hucMSCs could homing to damaged portal veins. Subsequent histological and immunohistochemical examinations demonstrated that intervention with miR-25-3p overexpression-modified hucMSCs significantly reduced damage and attenuated thrombosis in rat portal veins. The above findings indicate suggest that hucMSCs based on miR-25-3p modification may be a promising therapeutic approach for use in venous thrombotic diseases.


Sujet(s)
Prolifération cellulaire , Exosomes , Cellules endothéliales de la veine ombilicale humaine , Cellules souches mésenchymateuses , microARN , Veine porte , microARN/génétique , microARN/métabolisme , Humains , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Animaux , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Rats , Exosomes/métabolisme , Exosomes/génétique , Veine porte/métabolisme , Mouvement cellulaire/génétique , Rat Sprague-Dawley , Mâle , Thrombose veineuse/génétique , Thrombose veineuse/métabolisme , Thrombose veineuse/anatomopathologie , Thrombose veineuse/thérapie , Cellules cultivées , Techniques de coculture , Transduction du signal , Cordon ombilical/cytologie
16.
Arterioscler Thromb Vasc Biol ; 44(8): 1813-1832, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38957984

RÉSUMÉ

BACKGROUND: Cellular communication among different types of vascular cells is indispensable for maintaining vascular homeostasis and preventing atherosclerosis. However, the biological mechanism involved in cellular communication among these cells and whether this biological mechanism can be used to treat atherosclerosis remain unknown. We hypothesized that endothelial autophagy mediates the cellular communication in vascular tissue through exosome-mediated delivery of atherosclerosis-related genes. METHODS: Rapamycin and adeno-associated virus carrying Atg7 short hairpin RNA under the Tie (TEK receptor tyrosine kinase) promoter were used to activate and inhibit vascular endothelial autophagy in high-fat diet-fed ApoE-/- mice, respectively. miRNA microarray, in vivo and in vitro experiments, and human vascular tissue were used to explore the effects of endothelial autophagy on endothelial function and atherosclerosis and its molecular mechanisms. Quantitative polymerase chain reaction and miRNA sequencing were performed to determine changes in miRNA expression in exosomes. Immunofluorescence and exosome coculture experiments were conducted to examine the role of endothelial autophagy in regulating the communication between endothelial cells and smooth muscle cells (SMCs) via exosomal miRNA. RESULTS: Endothelial autophagy was inhibited in thoracic aortas of high-fat diet-fed ApoE-/- mice. Furthermore, rapamycin alleviated high-fat diet-induced atherosclerotic burden and endothelial dysfunction, while endothelial-specific Atg7 depletion aggravated the atherosclerotic burden. miRNA microarray, in vivo and in vitro experiments, and human vascular tissue analysis revealed that miR-204-5p was significantly increased in endothelial cells after high-fat diet exposure, which directly targeted Bcl2 to regulate endothelial cell apoptosis. Importantly, endothelial autophagy activation decreased excess miR-204-5p by loading miR-204-5p into multivesicular bodies and secreting it through exosomes. Moreover, exosomal miR-204-5p can effectively transport to SMCs, alleviating SMC calcification by regulating target proteins such as RUNX2 (runt-related transcription factor 2). CONCLUSIONS: Our study revealed the exosomal pathway by which endothelial autophagy protects atherosclerosis: endothelial autophagy activation transfers miR-204-5p from endothelial cells to SMCs via exosomes, both preventing endothelial apoptosis and alleviating SMC calcification. REGISTRATION: URL: https://www.chictr.org.cn/; Unique identifier: ChiCTR2200064155.


Sujet(s)
Athérosclérose , Autophagie , Communication cellulaire , Modèles animaux de maladie humaine , Exosomes , Souris de lignée C57BL , Souris invalidées pour les gènes ApoE , microARN , Myocytes du muscle lisse , microARN/métabolisme , microARN/génétique , Exosomes/métabolisme , Exosomes/génétique , Animaux , Athérosclérose/anatomopathologie , Athérosclérose/génétique , Athérosclérose/métabolisme , Athérosclérose/prévention et contrôle , Humains , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Mâle , Souris , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Cellules endothéliales de la veine ombilicale humaine/anatomopathologie , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , Protéine-7 associée à l'autophagie/métabolisme , Protéine-7 associée à l'autophagie/génétique , Cellules cultivées , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Plaque d'athérosclérose , Maladies de l'aorte/anatomopathologie , Maladies de l'aorte/génétique , Maladies de l'aorte/prévention et contrôle , Maladies de l'aorte/métabolisme , Techniques de coculture , Transduction du signal , Aorte thoracique/métabolisme , Aorte thoracique/anatomopathologie , Alimentation riche en graisse
17.
Biochem Pharmacol ; 226: 116413, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38971333

RÉSUMÉ

Chronic nonhealing diabetic wounds are a critical clinical challenge. Regulatory T cells (Tregs) are immunosuppressive modulators affecting wound healing progression by controlling the inflammatory response. The current study attempted to investigate whether the exosomes derived from cord blood (CB) Tregs can accelerate the healing process. Exosomes were isolated from CB-Treg cultures using ultracentrifugation and validated with different specific markers of exosomes. The purified CB-Treg-derived exosomes were co-cultured with peripheral blood mononuclear cells (PBMCs) and CD14+ monocytes. The migration-promoting effect of CB-Treg-derived exosomes on fibroblasts and endothelial cells was investigated. We used thermosensitive Pluronic F-127 hydrogel (PF-127) loaded with CB-Treg-derived exosomes in a diabetic wound healing mouse model. CB-Treg-derived exosomes with 30-120 nm diameters revealed exosome-specific markers, such as TSG101, Alix, and CD63. CB-Treg-derived exosomes were mainly bound to the monocytes when co-cultured with PBMCs, and promoted monocyte polarization to the anti-inflammatory phenotype (M2) in vitro. CB-Treg-derived exosomes enhanced the migration of endothelial cells and fibroblasts. Furthermore, CB-Treg-derived exosomes treatment accelerated wound healing by downregulating inflammatory factor levels and upregulating the M2 macrophage ratio in vivo. Our findings indicated that CB-Treg-derived exosomes could be a promising cell-free therapeutic strategy for diabetic wound healing, partly by targeting monocytes.


Sujet(s)
Diabète expérimental , Exosomes , Sang foetal , Monocytes , Lymphocytes T régulateurs , Cicatrisation de plaie , Exosomes/métabolisme , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Animaux , Cicatrisation de plaie/effets des médicaments et des substances chimiques , Cicatrisation de plaie/physiologie , Monocytes/métabolisme , Monocytes/effets des médicaments et des substances chimiques , Monocytes/immunologie , Souris , Sang foetal/cytologie , Humains , Diabète expérimental/métabolisme , Diabète expérimental/immunologie , Mâle , Souris de lignée C57BL , Techniques de coculture , Cellules cultivées , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques
18.
Biochem Pharmacol ; 226: 116414, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38972427

RÉSUMÉ

Lung adenocarcinoma (LUAD) is the most common histologic subtype of lung cancer. Angiogenesis plays a pivotal role in LUAD progression via supplying oxygen and nutrients for cancer cells. Non-coding miR-1293, a significantly up-regulated miRNA in LUAD tissues, can be potentially used as a novel biomarker for predicting the prognosis of LUAD patients. However, little information is available about the function of miR-1293 in LUAD progression especially cancer-induced angiogenesis. Herein, we found that miR-1293 knockdown could obviously attenuate LUAD-induced angiogenesis in vitro and down-regulate two most important pro-angiogenic cytokines VEGF-A and bFGF expression and secretion. Indeed, miR-1293 abrogation inactivated the angiogenesis-promoting ERK1/2 signaling characterized by decreased ERK1/2 phosphorylation and translocation from nucleus to cytoplasm. Next we found that miR-1293 knockdown reactivated the endogenous ERK1/2 pathway inhibitor Spry4 expression and Spry4 perturbance with specific siRNA transfection abolished the inhibition of ERK1/2 pathway and LUAD-induced angiogenesis by miR-1293 knockdown. Finally, with in vivo assay, we found obvious Spry4 up-regulation and VEGF-A, bFGF, ERK1/2 phosphorylation, micro-vessel density marker CD31 expression down-regulation in vivo, respectively. Collectively, these results indicated that miR-1293 knockdown could significantly attenuate LUAD angiogenesis via Spry4-mediated ERK1/2 signaling inhibition, which might be helpful for uncovering more functions of miR-1293 in LUAD and providing experimental basis for possible LUAD therapeutic strategy targeting miR-1293.


Sujet(s)
Adénocarcinome pulmonaire , Techniques de knock-down de gènes , Tumeurs du poumon , Système de signalisation des MAP kinases , microARN , Néovascularisation pathologique , Régulation positive , Humains , microARN/génétique , microARN/métabolisme , Néovascularisation pathologique/métabolisme , Néovascularisation pathologique/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Animaux , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/métabolisme , Système de signalisation des MAP kinases/physiologie , Système de signalisation des MAP kinases/génétique , Souris , Protéines de tissu nerveux/génétique , Protéines de tissu nerveux/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Protéines et peptides de signalisation intracellulaire/métabolisme , Souris nude , Mâle , Souris de lignée BALB C , Cellules A549 , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Femelle ,
19.
Curr Protein Pept Sci ; 25(7): 567-576, 2024.
Article de Anglais | MEDLINE | ID: mdl-39044556

RÉSUMÉ

BACKGROUND: Vascular Endothelial Growth Factor Receptors (VEGFR1 and VEGFR2) are tyrosine kinase receptors expressed on endothelial cells and tumor vessels and play an important role in angiogenesis. In this study, three repeats of VEGFR1 and VEGFR2 binding peptide (VGB3) were genetically fused to the truncated diphtheria toxin (TDT), and its in vitro activity was evaluated. METHODS: The recombinant construct (TDT-triVGB3) was expressed in bacteria cells and purified with nickel affinity chromatography. The binding capacity and affinity of TDT-triVGB3 were evaluated using the enzyme-linked immunosorbent assay. The inhibitory activity of TDT-triVGB3 on viability, migration, and tube formation of human endothelial cells was evaluated using MTT, migration, and tube formation assays. RESULTS: TDT-triVGB3 selectively detected VEGFR1 and VEGFR2 with high affinity in an enzyme- linked immunosorbent assay and significantly inhibited viability, migration, and tube formation of human endothelial cells. CONCLUSION: The developed TDT-triVGB3 is potentially a novel agent for targeting VEGFR1/ VEGFR2 over-expressing cancer cells.


Sujet(s)
Inhibiteurs de l'angiogenèse , Mouvement cellulaire , Toxine diphtérique , Cellules endothéliales de la veine ombilicale humaine , Récepteur-1 au facteur croissance endothéliale vasculaire , Récepteur-2 au facteur croissance endothéliale vasculaire , Humains , Récepteur-1 au facteur croissance endothéliale vasculaire/génétique , Récepteur-1 au facteur croissance endothéliale vasculaire/métabolisme , Récepteur-2 au facteur croissance endothéliale vasculaire/génétique , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme , Toxine diphtérique/génétique , Toxine diphtérique/pharmacologie , Toxine diphtérique/métabolisme , Mouvement cellulaire/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Inhibiteurs de l'angiogenèse/pharmacologie , Inhibiteurs de l'angiogenèse/génétique , Inhibiteurs de l'angiogenèse/composition chimique , Survie cellulaire/effets des médicaments et des substances chimiques , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/pharmacologie , Protéines de fusion recombinantes/métabolisme , Néovascularisation pathologique/génétique , Néovascularisation pathologique/métabolisme , Néovascularisation pathologique/anatomopathologie , Néovascularisation pathologique/traitement médicamenteux , Expression des gènes , Cellules endothéliales/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques
20.
Biochem Biophys Res Commun ; 727: 150321, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-38954982

RÉSUMÉ

Vascular endothelial growth factor (VEGF) is a pleiotropic growth factor that binds a broad spectrum of cell types and regulates diverse cellular processes, including angiogenesis, growth and survival. However, it is technically difficult to quantify VEGF-cell binding activity because of reversible nature of ligand-receptor interactions. Here we used T7 bacteriophage display to quantify and compare binding activity of three human VEGF-A (hVEGF) isoforms, including hVEGF111, 165 and 206. All three isoforms bound equally well to immobilized aflibercept, a decoy VEGF receptor. hVEGF111-Phage exhibited minimal binding to immobilized heparan sulfate, whereas hVEGF206-Phage and hVEGF165-Phage had the highest and intermediate binding to heparan, respectively. In vitro studies revealed that all three isoforms bound to human umbilical vein endothelial cells (HUVECs), HEK293 epithelial and SK-N-AS neuronal cells. hVEGF111-Phage has the lowest binding activity, while hVEGF206-Phage has the highest binding. hVEGF206-Phage was the most sensitive to detect VEGF-cell binding, albeit with the highest background binding to SK-N-AS cells. These results suggest that hVEGF206-Phage is the best-suited isoform to quantify VEGF-cell binding even though VEGF165 is the most biologically active. Furthermore, this study demonstrates the utility of T7 phage display as a platform for rapid and convenient ligand-cell binding quantification with pros and cons discussed.


Sujet(s)
Cellules endothéliales de la veine ombilicale humaine , Liaison aux protéines , Facteur de croissance endothéliale vasculaire de type A , Humains , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Cellules HEK293 , Isoformes de protéines/métabolisme , Récepteurs aux facteurs de croissance endothéliale vasculaire/métabolisme , Bactériophage T7/métabolisme , Bactériophage T7/génétique , Techniques d'exposition à la surface cellulaire/méthodes , Héparitine sulfate/métabolisme , Protéines de fusion recombinantes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE