Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 16.028
Filtrer
1.
J Biochem Mol Toxicol ; 38(8): e23785, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39051181

RÉSUMÉ

An arteriovenous fistula (AVF) is the preferred vascular access for hemodialysis in uremic patients, yet its dysfunction poses a significant clinical challenge. Venous stenosis, primarily caused by venous neointimal hyperplasia, is a key factor in the failure of vascular access. During vascular access dysfunction, endothelial cells (ECs) transform mechanical stimuli into intracellular signals and interact with vascular smooth muscle cells. Tanshinone IIA, an important compound derived from Salvia miltiorrhiza, has been widely used to treat cardiovascular diseases. However, its role in modulating ECs under uremic conditions remains incompletely understood. In this research, ECs were exposed to sodium tanshinone IIA sulfonate (STS) and subjected to shear stress and uremic conditions. The results indicate that STS can reduce the suppressive effects on the expression of NF-κB p65, JNK and Collagen I in uremia-induced ECs. Moreover, the downregulation of NF-κB p65, JNK and Collagen I can be enhanced through the inhibition of ERK1/2 and the upregulation of Caveolin-1. These findings suggest that tanshinone IIA may improve EC function under uremic conditions by targeting the Caveolin-1/ERK1/2 pathway, presenting tanshinone IIA as a potential therapeutic agent against AVF immaturity caused by EC dysfunction.


Sujet(s)
Abiétanes , Cavéoline-1 , Urémie , Urémie/métabolisme , Urémie/traitement médicamenteux , Urémie/anatomopathologie , Humains , Abiétanes/pharmacologie , Abiétanes/usage thérapeutique , Cavéoline-1/métabolisme , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Collagène de type I/métabolisme , Facteur de transcription RelA/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Phénanthrènes
2.
Int J Nanomedicine ; 19: 7123-7136, 2024.
Article de Anglais | MEDLINE | ID: mdl-39055375

RÉSUMÉ

Background: Drug therapy for eye diseases has been limited by multiple protective mechanisms of the eye, which can be improved using well-designed drug delivery systems. Mesoporous silica nanoparticles (MSNs) had been used in many studies as carriers of therapeutic agents for ocular diseases treatment. However, no studies have focused on ocular biosafety. Considering that MSNs containing tetrasulfur bonds have unique advantages and have drawn increasing attention in drug delivery systems, it is necessary to explore the ocular biosafety of tetrasulfur bonds before their widespread application as ophthalmic drug carriers. Methods: In this study, hollow mesoporous silica nanoparticles (HMSNs) with different tetrasulfur bond contents were prepared and characterized. The ocular biosafety of HMSN-E was evaluated in vitro on the three selected ocular cell lines, including corneal epithelial cells, lens epithelial cells and retinal endothelial cells (HREC), and in vivo by using topical eye drops and intravitreal injections. Results: In cellular experiments, HMSNs caused obvious S content-dependent cytotoxic effect. HMSNs with the highest tetrasulfur bond content (HMSN-E), showed the highest cytotoxicity among all the HMSNs, and HREC was the most vulnerable cell to HMSN-E. It was shown that HMSN-E could react with intracellular GSH to generate H2S and decrease intracellular GSH concentration. Treatment of HREC with HMSN-E increased intracellular ROS, decreased mitochondrial membrane potential, and induced cell cycle arrest at the G1/S checkpoint, finally caused apoptosis and necrosis of HREC. Topical eye drops of HMSN-E could cause corneal damage. The intravitreal injection of HMSN-E could induce inflammation in the vitreum and ganglion cell layers, resulting in vitreous opacities and retinal abnormalities. Conclusion: The incorporation of tetrasulfur bonds into HMSN can have toxic effects on ocular tissues. Therefore, when mesoporous silica nanocarriers are designed for ophthalmic pharmaceuticals, the ocular toxicity of the tetrasulfur bonds should be considered.


Sujet(s)
Nanoparticules , Silice , Humains , Animaux , Nanoparticules/composition chimique , Silice/composition chimique , Silice/toxicité , Lignée cellulaire , Porosité , Vecteurs de médicaments/composition chimique , Apoptose/effets des médicaments et des substances chimiques , Lapins , Survie cellulaire/effets des médicaments et des substances chimiques , Oeil/effets des médicaments et des substances chimiques , Solutions ophtalmiques/composition chimique , Solutions ophtalmiques/pharmacologie , Composés organiques du silicium/composition chimique , Composés organiques du silicium/toxicité , Espèces réactives de l'oxygène/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/effets des médicaments et des substances chimiques , Injections intravitréennes
3.
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
4.
Sci Rep ; 14(1): 16699, 2024 Jul 19.
Article de Anglais | MEDLINE | ID: mdl-39030268

RÉSUMÉ

To investigate the role of miR-223-3p in the modulatory effect of paeonol (Pae) on high glucose (HG)-induced endothelial cell apoptosis. HG (25 mmol/L) was used to induce cellular damage and apoptosis in the mouse cardiac microvascular endothelial cells (MCMECs). Various concentration of Pae was tested and 60 µmol/L Pae was selected for the subsequent studies. MCMECs were transfected with exogenous miR-223-3p mimics or anti-miR-223-3p inhibitors. Cell viability was assessed by MTT assay and apoptosis was quantified by flow cytometry. The expression of miR-223-3p and NLRP3 mRNA was measured using real-time quantitative RT-PCR, and protein level of NLRP3 and apoptosis-related proteins was detected by immunoblotting. Pae significantly attenuated HG-induced apoptosis of MCMECs in a concentration-dependent manner. In addition, Pae (60 µmol/L) significantly reversed HG-induced down-regulation of miR-223-3p and up-regulation of NLRP3. Pae (60 µmol/L) also significantly blocked HG-induced up-regulation of Bax and Caspase-3 as well as down-regulation of Bcl-2. Moreover, exogenous miR-223-3p mimics not only significantly attenuated HG-induced apoptosis, but also significantly suppressed NRLP-3 and pro-apoptotic proteins in the MCMECs. In contrast, transfection of exogenous miR-223-3p inhibitors into the MCMECs resulted in not only significantly increased apoptosis of the cells, but also significant suppression of NLRP3 and pro-apoptotic proteins in the cells. Pae attenuated HG-induced apoptosis of MCMECs in a concentration-dependent manner. MiR-223-3p may mediate the modulatory effects of Pae on MCMEC survival or apoptosis through targeting NLRP3 and regulating apoptosis-associated proteins.


Sujet(s)
Acétophénones , Apoptose , Cellules endothéliales , Glucose , microARN , Animaux , microARN/génétique , microARN/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Souris , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Glucose/pharmacologie , Acétophénones/pharmacologie , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Régulation positive/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Microvaisseaux/cytologie , Microvaisseaux/métabolisme , Microvaisseaux/effets des médicaments et des substances chimiques
6.
Drug Discov Ther ; 18(3): 199-206, 2024.
Article de Anglais | MEDLINE | ID: mdl-38987208

RÉSUMÉ

Senolytics are drugs that specifically target senescent cells. Flavonoids such as quercetin and fisetin possess selective senolytic activities. This study aims to investigate if chalcones exhibit anti-senescence activities. Anti-senescence effect of 11 chalcone derivatives on the replicative senescence human aortic endothelial cells (HAEC) and human fetal lung fibroblasts (IMR90) was evaluated. Compound 2 (4-methoxychalcone) and compound 4 (4-bromo-4'-methoxychalcone) demonstrated increased cytotoxicity in senescent HAEC compared to young HAEC, with significant differences on IC50 values. Their anti-senescence effects on HAEC exceeded fisetin. Higher selectivity of compound 4 toward HAEC over IMR90 could be attributed to 4-methoxy (4-OMe) substitution at ring A (R1). Chalcone derivatives have potentials as senolytics in mitigating replicative senescence, warranting further research and development on chalcones as anti-senescent agent.


Sujet(s)
Vieillissement de la cellule , Chalcones , Cellules endothéliales , Fibroblastes , Humains , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Cellules endothéliales/effets des médicaments et des substances chimiques , Chalcones/pharmacologie , Fibroblastes/effets des médicaments et des substances chimiques , Cellules cultivées , Sénothérapie/pharmacologie , Concentration inhibitrice 50 , Aorte/effets des médicaments et des substances chimiques , Aorte/cytologie , Relation structure-activité , Lignée cellulaire
7.
Int J Mol Sci ; 25(13)2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-39000380

RÉSUMÉ

Endothelial dysfunction often precedes the development of cardiovascular diseases, including heart failure. The cardioprotective benefits of sodium-glucose cotransporter 2 inhibitors (SGLT2is) could be explained by their favorable impact on the endothelium. In this review, we summarize the current knowledge on the direct in vitro effects of SGLT2is on endothelial cells, as well as the systematic observations in preclinical models. Four putative mechanisms are explored: oxidative stress, nitric oxide (NO)-mediated pathways, inflammation, and endothelial cell survival and proliferation. Both in vitro and in vivo studies suggest that SGLT2is share a class effect on attenuating reactive oxygen species (ROS) and on enhancing the NO bioavailability by increasing endothelial nitric oxide synthase activity and by reducing NO scavenging by ROS. Moreover, SGLT2is significantly suppress inflammation by preventing endothelial expression of adhesion receptors and pro-inflammatory chemokines in vivo, indicating another class effect for endothelial protection. However, in vitro studies have not consistently shown regulation of adhesion molecule expression by SGLT2is. While SGLT2is improve endothelial cell survival under cell death-inducing stimuli, their impact on angiogenesis remains uncertain. Further experimental studies are required to accurately determine the interplay among these mechanisms in various cardiovascular complications, including heart failure and acute myocardial infarction.


Sujet(s)
Inhibiteurs du cotransporteur sodium-glucose de type 2 , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Humains , Animaux , Stress oxydatif/effets des médicaments et des substances chimiques , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Monoxyde d'azote/métabolisme , Endothélium vasculaire/effets des médicaments et des substances chimiques , Endothélium vasculaire/métabolisme , Maladies cardiovasculaires/métabolisme , Maladies cardiovasculaires/traitement médicamenteux , Maladies cardiovasculaires/prévention et contrôle , Espèces réactives de l'oxygène/métabolisme , Nitric oxide synthase type III/métabolisme , Inflammation/métabolisme , Inflammation/traitement médicamenteux
8.
Molecules ; 29(13)2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38999055

RÉSUMÉ

Lignans, a class of secondary metabolites found in plants, along with their derivatives, exhibit diverse pharmacological activities, including antioxidant, antimicrobial, anti-inflammatory, and antiangiogenic ones. Angiogenesis, the formation of new blood vessels from pre-existing ones, is a crucial process for cancer growth and development. Several studies have elucidated the synergistic relationship between angiogenesis and inflammation in various inflammatory diseases, highlighting a correlation between inflammation and vascular endothelial growth factor (VEGF)-induced angiogenesis. Thus, the identification of novel molecules capable of modulating VEGF effects presents promising prospects for developing therapies aimed at stabilizing, reversing, or even arresting disease progression. Lignans often suffer from low aqueous solubility and, for their use, encapsulation in a delivery system is needed. In this research, a bioinspired benzoxantene has been encapsulated in solid lipid nanoparticles that have been characterized for their pharmacotechnical properties and their thermotropic behavior. The effects of these encapsulated nanoparticles on angiogenic parameters and inflammation in VEGF-induced angiogenesis were evaluated using human brain microvascular endothelial cells (HBMECs) as a human blood-brain barrier model.


Sujet(s)
Barrière hémato-encéphalique , Inflammation , Nanoparticules , Facteur de croissance endothéliale vasculaire de type A , Humains , Nanoparticules/composition chimique , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Barrière hémato-encéphalique/métabolisme , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Inflammation/anatomopathologie , Néovascularisation pathologique/traitement médicamenteux , Néovascularisation pathologique/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Lipides/composition chimique , Néovascularisation physiologique/effets des médicaments et des substances chimiques , , Liposomes
9.
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
10.
Int J Mol Sci ; 25(13)2024 Jun 22.
Article de Anglais | MEDLINE | ID: mdl-38999980

RÉSUMÉ

We have previously reported that, in aortic rings, 18:1 lysophosphatidic acid (LPA) can induce both vasodilation and vasoconstriction depending on the integrity of the endothelium. The predominant molecular species generated in blood serum are poly-unsaturated LPA species, yet the vascular effects of these species are largely unexplored. We aimed to compare the vasoactive effects of seven naturally occurring LPA species in order to elucidate their potential pathophysiological role in vasculopathies. Vascular tone was measured using myography, and thromboxane A2 (TXA2) release was detected by ELISA in C57Bl/6 mouse aortas. The Ca2+-responses to LPA-stimulated primary isolated endothelial cells were measured by Fluo-4 AM imaging. Our results indicate that saturated molecular species of LPA elicit no significant effect on the vascular tone of the aorta. In contrast, all 18 unsaturated carbon-containing (C18) LPAs (18:1, 18:2, 18:3) were effective, with 18:1 LPA being the most potent. However, following inhibition of cyclooxygenase (COX), these LPAs induced similar vasorelaxation, primarily indicating that the vasoconstrictor potency differed among these species. Indeed, C18 LPA evoked a similar Ca2+-signal in endothelial cells, whereas in endothelium-denuded aortas, the constrictor activity increased with the level of unsaturation, correlating with TXA2 release in intact aortas. COX inhibition abolished TXA2 release, and the C18 LPA induced vasoconstriction. In conclusion, polyunsaturated LPA have markedly increased TXA2-releasing and vasoconstrictor capacity, implying potential pathophysiological consequences in vasculopathies.


Sujet(s)
Aorte , Lysophospholipides , Souris de lignée C57BL , Thromboxane A2 , Vasoconstriction , Animaux , Thromboxane A2/métabolisme , Vasoconstriction/effets des médicaments et des substances chimiques , Souris , Lysophospholipides/métabolisme , Lysophospholipides/pharmacologie , Aorte/effets des médicaments et des substances chimiques , Aorte/métabolisme , Mâle , Cellules endothéliales/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , Vasodilatation/effets des médicaments et des substances chimiques , Calcium/métabolisme
11.
PeerJ ; 12: e17742, 2024.
Article de Anglais | MEDLINE | ID: mdl-39035169

RÉSUMÉ

Background: Allicin, a bioactive compound derived from garlic (Allium sativum), demonstrates antibacterial activity against a broad spectrum of bacteria including the most common meningitis pathogens. In order to advocate for allicin as a potential therapeutic candidate for bacterial meningitis, the present study aimed to assess the ability of allicin to cross the blood-brain barrier (BBB) using an in vitro model. Methods: The cell viability of the human brain endothelial cell line hCMEC/D3 after incubation with various concentrations of allicin was investigated using an MTT assay at 3 and 24 h. Additionally, reactive oxygen species (ROS) production of allicin-treated hCMEC/D3 cells was examined at 3 h. The concentrations of allicin that were not toxic to the cells, as determined by the MTT assay, and did not significantly increase ROS generation, were then used to investigate allicin's ability to traverse the in vitro BBB model for 3 h. High-performance liquid chromatography (HPLC) analysis was utilized to examine the allicin concentration capable of passing the in vitro BBB model. The cellular uptake experiments were subsequently performed to observe the uptake of allicin into hCMEC/D3 cells. The pkCSM online tool was used to predict the absorption, distribution, metabolism, excretion, and pharmacokinetic properties of allicin and S-allylmercaptoglutathione (GSSA). Results: The results from MTT assay indicated that the highest non-toxicity concentration of allicin on hCMEC/D3 cells was 5 µg/ml at 3 h and 2 µg/ml at 24 h. Allicin significantly enhanced ROS production of hCMEC/D3 cells at 10 µg/ml at 3 h. After applying the non-toxicity concentrations of allicin (0.5-5 µg/ml) to the in vitro BBB model for 3 h, allicin was not detectable in both apical and basolateral chambers in the presence of hCMEC/D3 cells. On the contrary, allicin was detected in both chambers in the absence of the cells. The results from cellular uptake experiments at 3 h revealed that hCMEC/D3 cells at 1 × 104 cells could uptake allicin at concentrations of 0.5, 1, and 2 µg/ml. Moreover, allicin uptake of hCMEC/D3 cells was proportional to the cell number, and the cells at 5 × 104 could completely uptake allicin at a concentration of 5 µg/ml within 0.5 h. The topological polar surface area (TPSA) predicting for allicin was determined to be 62.082 Å2, indicating its potential ability to cross the BBB. Additionally, the calculated logBB value surpassing 0.3 suggests that the compound may exhibit ease of penetration through the BBB. Conclusion: The present results suggested that allicin was rapidly taken up by hCMEC/D3 cells in vitro BBB model. The prediction results of allicin's distribution patterns suggested that the compound possesses the capability to enter the brain.


Sujet(s)
Barrière hémato-encéphalique , Survie cellulaire , Disulfures , Cellules endothéliales , Espèces réactives de l'oxygène , Acides sulfiniques , Acides sulfiniques/pharmacologie , Humains , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire , Chromatographie en phase liquide à haute performance
12.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 36(6): 664-668, 2024 Jun.
Article de Chinois | MEDLINE | ID: mdl-38991970

RÉSUMÉ

Resveratrol is a polyphenolic plant extract with many biological activities such as anti-inflammation and anti-oxidative stress. Vascular endothelial cell (VEC) is the main sites for maintaining normal vascular permeability and participating in vasomotor regulation and substance exchange. VEC injury plays a key role in various diseases or pathological processes such as cardiovascular disease, chronic inflammation and sepsis. Studies have shown that resveratrol protects VEC and reduces endothelial damage by regulating nitric oxide (NO) and its related enzymes, reducing oxidative stress and inhibiting apoptosis, thereby exerting beneficial effects.


Sujet(s)
Cellules endothéliales , Monoxyde d'azote , Resvératrol , Stilbènes , Resvératrol/pharmacologie , Humains , Cellules endothéliales/effets des médicaments et des substances chimiques , Stilbènes/pharmacologie , Monoxyde d'azote/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques
13.
PLoS One ; 19(7): e0306429, 2024.
Article de Anglais | MEDLINE | ID: mdl-38980867

RÉSUMÉ

Brucella abortus (Ba) is a pathogen that survives inside macrophages. Despite being its preferential niche, Ba infects other cells, as shown by the multiple signs and symptoms humans present. This pathogen can evade our immune system. Ba displays a mechanism of down-modulating MHC-I on monocytes/macrophages in the presence of IFN-γ (when Th1 response is triggered) without altering the total expression of MHC-I. The retained MHC-I proteins are located within the Golgi Apparatus (GA). The RNA of Ba is one of the PAMPs that trigger this phenomenon. However, we acknowledged whether this event could be triggered in other cells relevant during Ba infection. Here, we demonstrate that Ba RNA reduced the surface expression of MHC-I induced by IFN-γ in the human bronchial epithelium (Calu-6), the human alveolar epithelium (A-549) and the endothelial microvasculature (HMEC) cell lines. In Calu-6 and HMEC cells, Ba RNA induces the retention of MHC-I in the GA. This phenomenon was not observed in A-549 cells. We then evaluated the effect of Ba RNA on the secretion of IL-8, IL-6 and MCP-1, key cytokines in Ba infection. Contrary to our expectations, HMEC, Calu-6 and A-549 cells treated with Ba RNA had higher IL-8 and IL-6 levels compared to untreated cells. In addition, we showed that Ba RNA down-modulates the MHC-I surface expression induced by IFN-γ on human monocytes/macrophages via the pathway of the Epidermal Growth Factor Receptor (EGFR). So, cells were stimulated with an EGFR ligand-blocking antibody (Cetuximab) and Ba RNA. Neutralization of the EGFR to some extent reversed the down-modulation of MHC-I mediated by Ba RNA in HMEC and A-549 cells. In conclusion, this is the first study exploring a central immune evasion strategy, such as the downregulation of MHC-I surface expression, beyond monocytes and could shed light on how it persists effectively within the host, enduring unseen and escaping CD8+ T cell surveillance.


Sujet(s)
Brucella abortus , Cellules endothéliales , Cellules épithéliales , Antigènes d'histocompatibilité de classe I , Interféron gamma , Humains , Interféron gamma/métabolisme , Interféron gamma/pharmacologie , Cellules endothéliales/métabolisme , Cellules endothéliales/microbiologie , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/immunologie , Cellules épithéliales/métabolisme , Cellules épithéliales/microbiologie , Cellules épithéliales/immunologie , Antigènes d'histocompatibilité de classe I/métabolisme , Antigènes d'histocompatibilité de classe I/génétique , ARN bactérien/génétique , Lignée cellulaire , Régulation négative/effets des médicaments et des substances chimiques , Récepteurs ErbB/métabolisme , Brucellose/immunologie , Brucellose/métabolisme , Brucellose/microbiologie , Brucellose/génétique , Appareil de Golgi/métabolisme , Macrophages/métabolisme , Macrophages/immunologie , Macrophages/microbiologie , Monocytes/métabolisme , Monocytes/immunologie , Monocytes/effets des médicaments et des substances chimiques
14.
J Nanobiotechnology ; 22(1): 422, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-39014416

RÉSUMÉ

Vascularization plays a significant role in promoting the expedited process of bone regeneration while also enhancing the stability and viability of artificial bone implants. Although titanium alloy scaffolds were designed to mimic the porous structure of human bone tissues to facilitate vascularization in bone repair, their biological inertness restricted their broader utilization. The unique attribute of Metal-organic framework (MOF) MIL-53(Fe), known as "breathing", can facilitate the efficient adsorption of extracellular matrix proteins and thus provide the possibility for efficient interaction between scaffolds and cell adhesion molecules, which helps improve the bioactivity of the titanium alloy scaffolds. In this study, MIL-53(Fe) was synthesized in situ on the scaffold after hydrothermal treatment. The MIL-53(Fe) endowed the scaffold with superior protein absorption ability and preferable biocompatibility. The scaffolds have been shown to possess favorable osteogenesis and angiogenesis inducibility. It was indicated that MIL-53(Fe) modulated the mechanotransduction process of endothelial cells and induced increased cell stiffness by promoting the adsorption of adhesion-mediating extracellular matrix proteins to the scaffold, such as laminin, fibronectin, and perlecan et al., which contributed to the activation of the endothelial tip cell phenotype at sprouting angiogenesis. Therefore, this study effectively leveraged the intrinsic "breathing" properties of MIL-53 (Fe) to enhance the interaction between titanium alloy scaffolds and vascular endothelial cells, thereby facilitating the vascularization inducibility of the scaffold, particularly during the sprouting angiogenesis phase. This study indicates that MIL-53(Fe) coating represents a promising strategy to facilitate accelerated and sufficient vascularization and uncovers the scaffold-vessel interaction from a biomechanical perspective.


Sujet(s)
Néovascularisation physiologique , Structures d'échafaudage tissulaires , Titane , Titane/composition chimique , Humains , Structures d'échafaudage tissulaires/composition chimique , Néovascularisation physiologique/effets des médicaments et des substances chimiques , Cellules endothéliales/effets des médicaments et des substances chimiques , Réseaux organométalliques/composition chimique , Réseaux organométalliques/pharmacologie , Matériaux biocompatibles/composition chimique , Matériaux biocompatibles/pharmacologie , Ostéogenèse/effets des médicaments et des substances chimiques , Alliages/composition chimique , Cellules endothéliales de la veine ombilicale humaine , Prothèses et implants , Mécanotransduction cellulaire , Adhérence cellulaire/effets des médicaments et des substances chimiques , Ingénierie tissulaire/méthodes
15.
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
16.
J Transl Med ; 22(1): 672, 2024 Jul 20.
Article de Anglais | MEDLINE | ID: mdl-39033271

RÉSUMÉ

BACKGROUND: T cells play a pivotal role in chemotherapy-triggered anti-tumor effects. Emerging evidence underscores the link between impaired anti-tumor immune responses and resistance to paclitaxel therapy in triple-negative breast cancer (TNBC). Tumor-related endothelial cells (ECs) have potential immunoregulatory activity. However, how ECs regulate T cell activity during TNBC chemotherapy remains poorly understood. METHODS: Single-cell analysis of ECs in patients with TNBC receiving paclitaxel therapy was performed using an accessible single-cell RNA sequencing (scRNA-seq) dataset to identify key EC subtypes and their immune characteristics. An integrated analysis of a tumor-bearing mouse model, immunofluorescence, and a spatial transcriptome dataset revealed the spatial relationship between ECs, especially Tumor necrosis factor receptor (TNFR) 2+ ECs, and CD8+ T cells. RNA sequencing, CD8+ T cell proliferation assays, flow cytometry, and bioinformatic analyses were performed to explore the immunosuppressive function of TNFR2 in ECs. The downstream metabolic mechanism of TNFR2 was further investigated using RNA sequencing, cellular glycolysis assays, and western blotting. RESULTS: In this study, we identified an immunoregulatory EC subtype, characterized by enhanced TNFR2 expression in non-responders. By a mouse model of TNBC, we revealed a dynamic reduction in the proportion of the CD8+ T cell-contacting tumor vessels that could co-localize spatially with CD8+ T cells during chemotherapy and an increased expression of TNFR2 by ECs. TNFR2 suppresses glycolytic activity in ECs by activating NF-κB signaling in vitro. Tuning endothelial glycolysis enhances programmed death-ligand (PD-L) 1-dependent inhibitory capacity, thereby inducing CD8+ T cell suppression. In addition, TNFR2+ ECs showed a greater spatial affinity for exhausted CD8+ T cells than for non-exhausted CD8+ T cells. TNFR2 blockade restores impaired anti-tumor immunity in vivo, leading to the loss of PD-L1 expression by ECs and enhancement of CD8+ T cell infiltration into the tumors. CONCLUSIONS: These findings reveal the suppression of CD8+ T cells by ECs in chemoresistance and indicate the critical role of TNFR2 in driving the immunosuppressive capacity of ECs via tuning glycolysis. Targeting endothelial TNFR2 may serve as a potent strategy for treating TNBC with paclitaxel.


Sujet(s)
Lymphocytes T CD8+ , Résistance aux médicaments antinéoplasiques , Cellules endothéliales , Glycolyse , Récepteur au facteur de nécrose tumorale de type II , Tumeurs du sein triple-négatives , Récepteur au facteur de nécrose tumorale de type II/métabolisme , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Glycolyse/effets des médicaments et des substances chimiques , Animaux , Humains , Cellules endothéliales/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , Femelle , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/immunologie , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/métabolisme , Lignée cellulaire tumorale , Paclitaxel/pharmacologie , Paclitaxel/usage thérapeutique , Souris , Transduction du signal/effets des médicaments et des substances chimiques
17.
J Nanobiotechnology ; 22(1): 440, 2024 Jul 27.
Article de Anglais | MEDLINE | ID: mdl-39061065

RÉSUMÉ

Inflammatory factors and reactive oxygen species (ROS) are risk factors for atherosclerosis. Many existing therapies use ROS-sensitive delivery systems to alleviate atherosclerosis, which achieved certain efficacy, but cannot eliminate excessive ROS. Moreover, the potential biological safety concerns of carrier materials through chemical synthesis cannot be ignored. Herein, an amphiphilic low molecular weight heparin- lipoic acid conjugate (LMWH-LA) was used as a ROS-sensitive carrier material, which consisted of injectable drug molecules used clinically, avoiding unknown side effects. LMWH-LA and curcumin (Cur) self-assembled to form LLC nanoparticles (LLC NPs) with LMWH as shell and LA/Cur as core, in which LMWH could target P-selectin on plaque endothelial cells and competitively block the migration of monocytes to endothelial cells to inhibit the origin of ROS and inflammatory factors, and LA could be oxidized to trigger hydrophilic-hydrophobic transformation and accelerate the release of Cur. Cur released within plaques further exerted anti-inflammatory and antioxidant effects, thereby suppressing ROS and inflammatory factors. We used ultrasound imaging, pathology and serum analysis to evaluate the therapeutic effect of nanoparticles on atherosclerotic plaques in apoe-/- mice, and the results showed that LLC showed significant anti-atherosclerotic effects. Our finding provided a promising therapeutic nanomedicine for the treatment of atherosclerosis.


Sujet(s)
Anti-inflammatoires , Athérosclérose , Curcumine , Nanoparticules , Plaque d'athérosclérose , Espèces réactives de l'oxygène , Animaux , Espèces réactives de l'oxygène/métabolisme , Souris , Curcumine/pharmacologie , Curcumine/composition chimique , Athérosclérose/traitement médicamenteux , Nanoparticules/composition chimique , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/composition chimique , Humains , Plaque d'athérosclérose/traitement médicamenteux , Acide lipoïque/composition chimique , Acide lipoïque/pharmacologie , Héparine bas poids moléculaire/pharmacologie , Héparine bas poids moléculaire/composition chimique , Héparine bas poids moléculaire/usage thérapeutique , Souris de lignée C57BL , Inflammation/traitement médicamenteux , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Mâle , Sélectine P/métabolisme , Vecteurs de médicaments/composition chimique , Antioxydants/pharmacologie , Antioxydants/composition chimique
18.
Eur J Pharm Sci ; 200: 106847, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38972611

RÉSUMÉ

Exogenous insulin-like growth factor-1 (IGF-1) has been reported to promote wound healing through regulation of vascular endothelial cells (VECs). Despite the existing studies of IGF-1 on VEC and its role in angiogenesis, the mechanisms regarding anti-inflammatory and angiogenetic effects of IGF-1 remain unclear. In this study, we investigated the wound-healing process and the related signaling pathway of IGF-1 using an inflammation model induced by IFN-γ. The results demonstrated that IGF-1 can increase cell proliferation, suppress inflammation in VECs, and promote angiogenesis. In vivo studies further confirmed that IGF-1 can reduce inflammation, enhance vascular regeneration, and improve re-epithelialization and collagen deposition in acute wounds. Importantly, the Ras/PI3K/IKK/NF-κB signaling pathways was identified as the mechanisms through which IGF-1 exerts its anti-inflammatory and pro-angiogenic effects. These findings contribute to the understanding of IGF-1's role in wound healing and may have implications for the development of new wound treatment approaches.


Sujet(s)
Inflammation , Facteur de croissance IGF-I , Facteur de transcription NF-kappa B , Transduction du signal , Cicatrisation de plaie , Facteur de croissance IGF-I/métabolisme , Animaux , Cicatrisation de plaie/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Inflammation/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Humains , Néovascularisation physiologique/effets des médicaments et des substances chimiques , Protéines G ras/métabolisme , Mâle , I-kappa B Kinase/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris , Souris de lignée C57BL , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Anti-inflammatoires/pharmacologie , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Interféron gamma/métabolisme , Interféron gamma/pharmacologie ,
19.
Sci Rep ; 14(1): 15178, 2024 07 10.
Article de Anglais | MEDLINE | ID: mdl-38987553

RÉSUMÉ

The evolution of endovascular therapies, particularly in the field of intracranial aneurysm treatment, has been truly remarkable and is characterized by the development of various stents. However, ischemic complications related to thrombosis or downstream emboli pose a challenge for the broader clinical application of such stents. Despite advancements in surface modification technologies, an ideal coating that fulfills all the desired requirements, including anti-thrombogenicity and swift endothelialization, has not been available. To address these issues, we investigated a new coating comprising 3-aminopropyltriethoxysilane (APTES) with both anti-thrombogenic and cell-adhesion properties. We assessed the anti-thrombogenic property of the coating using an in vitro blood loop model by evaluating the platelet count and the level of the thrombin-antithrombin (TAT) complex, and investigating thrombus formation on the surface using scanning electron microscopy (SEM). We then assessed endothelial cell adhesion on the metal surfaces. In vitro blood tests revealed that, compared to a bare stent, the coating significantly inhibited platelet reduction and thrombus formation; more human serum albumin spontaneously adhered to the coated surface to block thrombogenic activation in the blood. Cell adhesion tests also indicated a significant increase in the number of cells adhering to the APTES-coated surfaces compared to the numbers adhering to either the bare stent or the stent coated with an anti-fouling phospholipid polymer. Finally, we performed an in vivo safety test by implanting coated stents into the internal thoracic arteries and ascending pharyngeal arteries of minipigs, and subsequently assessing the health status and vessel patency of the arteries by angiography over the course of 1 week. We found that there were no adverse effects on the pigs and the vascular lumens of their vessels were well maintained in the group with APTES-coated stents. Therefore, our new coating exhibited both high anti-thrombogenicity and cell-adhesion properties, which fulfill the requirements of an implantable stent.


Sujet(s)
Adhérence cellulaire , Matériaux revêtus, biocompatibles , Propylamines , Silanes , Endoprothèses , Thrombose , Silanes/composition chimique , Silanes/pharmacologie , Animaux , Adhérence cellulaire/effets des médicaments et des substances chimiques , Humains , Endoprothèses/effets indésirables , Suidae , Matériaux revêtus, biocompatibles/composition chimique , Matériaux revêtus, biocompatibles/pharmacologie , Propylamines/pharmacologie , Propylamines/composition chimique , Adsorption , Thrombose/prévention et contrôle , Fibrinolytiques/pharmacologie , Fibrinolytiques/composition chimique , Plaquettes/effets des médicaments et des substances chimiques , Plaquettes/métabolisme , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme
20.
Food Funct ; 15(15): 7849-7864, 2024 Jul 29.
Article de Anglais | MEDLINE | ID: mdl-38962816

RÉSUMÉ

As the first line of defense, the skin is equipped with various physiological mechanisms positioned to prevent incoming oxidative damage from numerous environmental insults. With persistent exposure to the environment, understanding ways to augment the skin defenses is paramount in protecting from premature aging. In this study, we investigated the ability of five dietary phenolic metabolites, typically found in the bloodstream after wild blueberry consumption, to successfully defend the skin from UV light exposure in a novel ex vivo co-culture model of human skin explants and primary endothelial cells. Skin explants, placed in transwell inserts, were exposed to UV, and subsequently co-cultured with endothelial cells. When the endothelial cells had been pretreated with the bioactive metabolites at physiological concentrations (hippuric acid 3000 nM, isoferulic acid 1000 nM, salicylic acid 130 nM, benzoic acid 900 nM, α-hydroxyhippuric acid 400 nM) cutaneous damage was prevented on the co-cultured with UV-challenged skin explants. Co-culture with non-pretreated endothelial cells did not protect skin explants. Specifically, the pretreatment was able to reduce skin lipid peroxidation (measured as 4-hydroxynonenal protein adducts), and pro-inflammatory enzymes such as cyclooxygenase 2 (COX-2) and NADPH oxidase 4 (NOX-4). Furthermore, pretreatment with the metabolites prevented UV-induced release of inflammatory cytokines such as IL-1ß and IL-8 as well as nitric oxides (NO) levels. In addition, the metabolites showed an impressive ability to prevent the loss of cutaneous structural proteins including involucrin and collagen type 1. Of note, endothelial cells cultured with UV exposed skin explants exhibited increased oxidative stress demonstrated by heme oxygenase-1 (HO-1) up-regulation which was significantly prevented in the metabolite treated models. These findings highlight the ability of dietary polyphenolic metabolites to improve cutaneous defenses against extrinsic stressors.


Sujet(s)
Myrtillier , Cellules endothéliales , Hydroxybenzoates , Peau , Rayons ultraviolets , Humains , Myrtillier/composition chimique , Peau/métabolisme , Peau/effets des médicaments et des substances chimiques , Hydroxybenzoates/pharmacologie , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Techniques de coculture , Peroxydation lipidique/effets des médicaments et des substances chimiques , Interleukine-8/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE