Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.729
Filtrer
1.
J Am Heart Assoc ; 13(12): e032971, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38842271

RÉSUMÉ

BACKGROUND: The mineralocorticoid receptor plays a significant role in the development of chronic kidney disease (CKD) and associated cardiovascular complications. Classic steroidal mineralocorticoid receptor antagonists are a therapeutic option, but their use in the clinic is limited due to the associated risk of hyperkalemia in patients with CKD. Finerenone is a nonsteroidal mineralocorticoid receptor antagonist that has been recently investigated in 2 large phase III clinical trials (FIDELIO-DKD [Finerenone in Reducing Kidney Failure and Disease Progression in Diabetic Kidney Disease] and FIGARO-DKD [Finerenone in Reducing Cardiovascular Mortality and Morbidity in Diabetic Kidney Disease]), showing reductions in kidney and cardiovascular outcomes. METHODS AND RESULTS: We tested whether finerenone improves renal and cardiac function in a preclinical nondiabetic CKD model. Twelve weeks after 5/6 nephrectomy, the rats showed classic signs of CKD characterized by a reduced glomerular filtration rate and increased kidney weight, associated with left ventricular (LV) diastolic dysfunction and decreased LV perfusion. These changes were associated with increased cardiac fibrosis and reduced endothelial nitric oxide synthase activating phosphorylation (ser 1177). Treatment with finerenone prevented LV diastolic dysfunction and increased LV tissue perfusion associated with a reduction in cardiac fibrosis and increased endothelial nitric oxide synthase phosphorylation. Curative treatment with finerenone improves nondiabetic CKD-related LV diastolic function associated with a reduction in cardiac fibrosis and increased cardiac phosphorylated endothelial nitric oxide synthase independently from changes in kidney function. Short-term finerenone treatment decreased LV end-diastolic pressure volume relationship and increased phosphorylated endothelial nitric oxide synthase and nitric oxide synthase activity. CONCLUSIONS: We showed that the nonsteroidal mineralocorticoid receptor antagonist finerenone reduces renal hypertrophy and albuminuria, attenuates cardiac diastolic dysfunction and cardiac fibrosis, and improves cardiac perfusion in a preclinical nondiabetic CKD model.


Sujet(s)
Modèles animaux de maladie humaine , Fibrose , Antagonistes des récepteurs des minéralocorticoïdes , Naphtyridines , Nitric oxide synthase type III , Insuffisance rénale chronique , Dysfonction ventriculaire gauche , Animaux , Antagonistes des récepteurs des minéralocorticoïdes/pharmacologie , Antagonistes des récepteurs des minéralocorticoïdes/usage thérapeutique , Insuffisance rénale chronique/traitement médicamenteux , Insuffisance rénale chronique/physiopathologie , Insuffisance rénale chronique/complications , Insuffisance rénale chronique/métabolisme , Naphtyridines/pharmacologie , Naphtyridines/usage thérapeutique , Dysfonction ventriculaire gauche/physiopathologie , Dysfonction ventriculaire gauche/traitement médicamenteux , Dysfonction ventriculaire gauche/étiologie , Dysfonction ventriculaire gauche/métabolisme , Mâle , Nitric oxide synthase type III/métabolisme , Débit de filtration glomérulaire/effets des médicaments et des substances chimiques , Fonction ventriculaire gauche/effets des médicaments et des substances chimiques , Diastole/effets des médicaments et des substances chimiques , Rein/effets des médicaments et des substances chimiques , Rein/physiopathologie , Rein/métabolisme , Phosphorylation , Myocarde/métabolisme , Myocarde/anatomopathologie , Rat Sprague-Dawley , Rats , Néphrectomie
2.
Int Immunopharmacol ; 136: 112410, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-38843641

RÉSUMÉ

Impaired wound healing in diabetes results from a complex interplay of factors that disrupt epithelialization and wound closure. MG53, a tripartite motif (TRIM) family protein, plays a key role in repairing cell membrane damage and facilitating tissue regeneration. In this study, bone marrow-derived mesenchymal stem cells (BMSCs) were transduced with lentiviral vectors overexpressing MG53 to investigate their efficacy in diabetic wound healing. Using a db/db mouse wound model, we observed that BMSCs-MG53 significantly enhanced diabetic wound healing. This improvement was associated with marked increase in re-epithelialization and vascularization. BMSCs-MG53 promoted recruitment and survival of BMSCs, as evidenced by an increase in MG53/Ki67-positive BMSCs and their improved response to scratch wounding. The combination therapy also promoted angiogenesis in diabetic wound tissues by upregulating the expression of angiogenic growth factors. MG53 overexpression accelerated the differentiation of BMSCs into endothelial cells, manifested as the formation of mature vascular network structure and a remarkable increase in DiI-Ac-LDL uptake. Our mechanistic investigation revealed that MG53 binds to caveolin-3 (CAV3) and subsequently increases phosphorylation of eNOS, thereby activating eNOS/NO signaling. Notably, CAV3 knockdown reversed the promoting effects of MG53 on BMSCs endothelial differentiation. Overall, our findings support the notion that MG53 binds to CAV3, activates eNOS/NO signaling pathway, and accelerates the therapeutic effect of BMSCs in the context of diabetic wound healing. These insights hold promise for the development of innovative strategies for treating diabetic-related impairments in wound healing.


Sujet(s)
Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Nitric oxide synthase type III , Monoxyde d'azote , Transduction du signal , Cicatrisation de plaie , Animaux , Cellules souches mésenchymateuses/métabolisme , Nitric oxide synthase type III/métabolisme , Souris , Monoxyde d'azote/métabolisme , Mâle , Souris de lignée C57BL , Néovascularisation physiologique , Cellules cultivées , Humains , Diabète expérimental/thérapie , Diabète expérimental/métabolisme , Différenciation cellulaire , Protéines membranaires
3.
Pharmazie ; 79(6): 101-108, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38877681

RÉSUMÉ

In this study, we hypothesized that lixisenatide (LIX) and ticagrelor (TIC) could have a protective effect against type 2 diabetes mellitus (T2DM)-induced vascular damage. Furthermore, we explored the possible additional protective effect of co-administering LIX and TIC in the treatment regimen. Methods: 50 male rats were divided into five groups, each comprising 10 rats: C (control), D (T2DM rats), D + LIX (T2DM rats treated with LIX for 4 weeks), D + TIC (T2DM rats treated with TIC for 4 weeks), and D + LIX + TIC (T2DM rats treated with LIX + TIC for 4 weeks). Results: The D group showed an increase in body weight, blood glucose, hemostatic model assessment for insulin resistance (HOMA-IR), aorta reactive oxygen species (ROS), and nuclear factor kappa B (NF-κ B), along with a reduction in serum insulin, aorta superoxide dismutase (SOD), glutathione reduced (GSH), nuclear factor erythroid-2 (NrF2), hemeoxygenase-1 (HO-1), and endothelial nitric oxide synthase (eNOS). Deterioration in the aorta histopathological condition, coupled with a noticeable impairment in vascular reactivity compared to the C group, was observed. A single administration of LIX showed a reduction in body weight, blood glucose, HOMA-IR, aorta ROS, and NF-κ B, accompanied by an increase in serum insulin, aorta SOD, GSH, NrF2, HO-1, and eNOS. Amelioration in the aorta histopathological condition and improved vascular reactivity compared to the D group were reported. Similarly, a single administration of TIC showed a reduction in aorta ROS and NF-κ B, along with an increase in aorta SOD, GSH, NrF2, HO-1, and eNOS. A slight amelioration was detected in the aorta histopathological condition, with improved vascular reactivity compared to the D group. The combined administration of LIX and TIC showed a reduction in aorta ROS and NF-κ B, along with an increase in aorta GSH, SOD, HO-1, and eNOS. This was combined with evident amelioration in the aorta histopathological condition and noticeable improvement in vascular reactivity compared to the single treatment with either LIX or TIC group. Conclusion: The present study introduces clear evidence that the administration of LIX and TIC can improve metabolic and vascular complications of T2DM through modulating eNOS and NrF2 /HO-1 signaling. The combined administration of LIX and TIC produced more significant effects than a single treatment.


Sujet(s)
Diabète expérimental , Facteur-2 apparenté à NF-E2 , Nitric oxide synthase type III , Peptides , Espèces réactives de l'oxygène , Transduction du signal , Ticagrélor , Animaux , Mâle , Nitric oxide synthase type III/métabolisme , Rats , Transduction du signal/effets des médicaments et des substances chimiques , Ticagrélor/pharmacologie , Ticagrélor/administration et posologie , Peptides/pharmacologie , Peptides/administration et posologie , Facteur-2 apparenté à NF-E2/métabolisme , Diabète expérimental/traitement médicamenteux , Diabète expérimental/complications , Espèces réactives de l'oxygène/métabolisme , Glycémie/effets des médicaments et des substances chimiques , Insulinorésistance , Diabète de type 2/traitement médicamenteux , Diabète de type 2/complications , Diabète de type 2/métabolisme , Rat Sprague-Dawley , Heme oxygenase (decyclizing)/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Hypoglycémiants/pharmacologie , Hypoglycémiants/administration et posologie , Heme oxygenase-1/métabolisme , Insuline , Stress oxydatif/effets des médicaments et des substances chimiques , Superoxide dismutase/métabolisme , Synergie des médicaments , Récepteur du peptide-2 similaire au glucagon
4.
J Cardiothorac Surg ; 19(1): 312, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38824570

RÉSUMÉ

OBJECTIVE: About 10% of patients after cardiopulmonary bypass (CPB) would undergo acute liver injury, which aggravated the mortality of patients. Ac2-26 has been demonstrated to ameliorate organic injury by inhibiting inflammation. The present study aims to evaluate the effect and mechanism of Ac2-26 on acute liver injury after CPB. METHODS: A total of 32 SD rats were randomized into sham, CPB, Ac, and Ac/AKT1 groups. The rats only received anesthesia, and rats in other groups received CPB. The rats in Ac/AKT1 were pre-injected with the shRNA to interfere with the expression of AKT1. The rats in CPB were injected with saline, and rats in Ac and Ac/AKT1 groups were injected with Ac2-26. After 12 h of CPB, all the rats were sacrificed and the peripheral blood and liver samples were collected to analyze. The inflammatory factors in serum and liver were detected. The liver function was tested, and the pathological injury of liver tissue was evaluated. RESULTS: Compared with the sham group, the inflammatory factors, liver function, and pathological injury were worsened after CPB. Compared with the CPB group, the Ac2-26 significantly decreased the pro-inflammatory factors and increased the anti-inflammatory factor, improved liver function, and ameliorated the pathological injury. All the therapeutic effects of Ac2-26 were notably attenuated by the shRNA of AKT1. The Ac2-26 increased the GSK3ß and eNOS, and this promotion was inhibited by the shRNA. CONCLUSION: The Ac2-26 significantly treated the liver injury, inhibited inflammation, and improved liver function. The effect of Ac2-26 on liver injury induced by CPB was partly associated with the promotion of AKT1/GSK3ß/eNOS.


Sujet(s)
Pontage cardiopulmonaire , Glycogen synthase kinase 3 beta , Nitric oxide synthase type III , Protéines proto-oncogènes c-akt , Rat Sprague-Dawley , Animaux , Pontage cardiopulmonaire/effets indésirables , Protéines proto-oncogènes c-akt/métabolisme , Glycogen synthase kinase 3 beta/métabolisme , Rats , Nitric oxide synthase type III/métabolisme , Mâle , Modèles animaux de maladie humaine , Foie/anatomopathologie , Transduction du signal
5.
Int J Mol Med ; 54(1)2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38874017

RÉSUMÉ

In paraquat (PQ)­induced acute lung injury (ALI)/ acute respiratory distress syndrome, PQ disrupts endothelial cell function and vascular integrity, which leads to increased pulmonary leakage. Anthrahydroquinone­2,6­disulfonate (AH2QDS) is a reducing agent that attenuates the extent of renal injury and improves survival in PQ­intoxicated Sprague­Dawley (SD) rats. The present study aimed to explore the beneficial role of AH2QDS in PQ­induced ALI and its related mechanisms. A PQ­intoxicated ALI model was established using PQ gavage in SD rats. Human pulmonary microvascular endothelial cells (HPMECs) were challenged with PQ. Superoxide dismutase, malondialdehyde, reactive oxygen species and nitric oxide (NO) fluorescence were examined to detect the level of oxidative stress in HPMECs. The levels of TNF­α, IL­1ß and IL­6 were assessed using an ELISA. Transwell and Cell Counting Kit­8 assays were performed to detect the migration and proliferation of the cells. The pathological changes in lung tissues and blood vessels were examined by haematoxylin and eosin staining. Evans blue staining was used to detect pulmonary microvascular permeability. Western blotting was performed to detect target protein levels. Immunofluorescence and immunohistochemical staining were used to detect the expression levels of target proteins in HPMECs and lung tissues. AH2QDS inhibited inflammatory responses in lung tissues and HPMECs, and promoted the proliferation and migration of HPMECs. In addition, AH2QDS reduced pulmonary microvascular permeability by upregulating the levels of vascular endothelial­cadherin, zonula occludens­1 and CD31, thereby attenuating pathological changes in the lungs in rats. Finally, these effects may be related to the suppression of the phosphatidylinositol­3­kinase (PI3K)/protein kinase B (AKT)/endothelial­type NO synthase (eNOS) signalling pathway in endothelial cells. In conclusion, AH2QDS ameliorated PQ­induced ALI by improving alveolar endothelial barrier disruption via modulation of the PI3K/AKT/eNOS signalling pathway, which may be an effective candidate for the treatment of PQ­induced ALI.


Sujet(s)
Lésion pulmonaire aigüe , Perméabilité capillaire , Poumon , Nitric oxide synthase type III , Paraquat , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Rat Sprague-Dawley , Transduction du signal , Animaux , Lésion pulmonaire aigüe/métabolisme , Lésion pulmonaire aigüe/traitement médicamenteux , Lésion pulmonaire aigüe/induit chimiquement , Lésion pulmonaire aigüe/anatomopathologie , Protéines proto-oncogènes c-akt/métabolisme , Nitric oxide synthase type III/métabolisme , Perméabilité capillaire/effets des médicaments et des substances chimiques , Phosphatidylinositol 3-kinases/métabolisme , Humains , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/métabolisme , Poumon/effets des médicaments et des substances chimiques , Paraquat/effets indésirables , Paraquat/toxicité , Rats , Cellules endothéliales/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , Stress oxydatif/effets des médicaments et des substances chimiques
6.
Life Sci ; 351: 122862, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38917872

RÉSUMÉ

The primary and initial manifestations of hypertension encompass arterial hypoelasticity and histiocyte senescence. Oxidative stress plays a pivotal role in the progression of senescence. Elevated intracellular oxidative stress levels will directly induce cell damage, disrupt normal physiological signal transduction, which can cause mitochondrial dysfunction to accelerate the process of senescence. Alizarin, an anthraquinone active ingredient isolated from Rubia cordifolia L., has a variety of pharmacological effects, including antioxidant, anti-inflammatory and anti-platelet. Nevertheless, its potential in lowering blood pressure (BP) and mitigating hypertension-induced vascular senescence remains uncertain. In this study, we used spontaneously hypertensive rats (SHR) and human umbilical vein endothelial cells (HUVECs) to establish a model of vascular senescence in hypertension. Our aim was to elucidate the mechanisms underpinning the vascular protective effects of Alizarin. By assessing systolic blood pressure (SBP) and diastolic blood pressure (DBP), H&E staining, SA-ß-Gal staining, vascular function, oxidative stress levels, calcium ion concentration and mitochondrial membrane potential, we found that Alizarin not only restored SBP and increased endothelium-dependent relaxation (EDR) in SHR, but also inhibited oxidative stress-induced mitochondrial damage and significantly delayed the vascular senescence effect in hypertension, and the mechanism may be related to the activation of VEGFR2/eNOS signaling pathway.


Sujet(s)
Anthraquinones , Antihypertenseurs , Vieillissement de la cellule , Cellules endothéliales de la veine ombilicale humaine , Hypertension artérielle , Mitochondries , Nitric oxide synthase type III , Stress oxydatif , Rats de lignée SHR , Transduction du signal , Récepteur-2 au facteur croissance endothéliale vasculaire , Stress oxydatif/effets des médicaments et des substances chimiques , Animaux , Humains , Rats , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Anthraquinones/pharmacologie , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Antihypertenseurs/pharmacologie , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Nitric oxide synthase type III/métabolisme , Hypertension artérielle/métabolisme , Hypertension artérielle/traitement médicamenteux , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Mâle , Pression sanguine/effets des médicaments et des substances chimiques , Rats de lignée WKY
7.
Int J Mol Sci ; 25(12)2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38928382

RÉSUMÉ

Graphene, when electrified, generates far-infrared radiation within the wavelength range of 4 µm to 14 µm. This range closely aligns with the far-infrared band (3 µm to 15 µm), which produces unique physiological effects. Contraction and relaxation of vascular smooth muscle play a significant role in primary hypertension, involving the nitric oxide-soluble guanylate cyclase-cyclic guanosine monophosphate pathway and the renin-angiotensin-aldosterone system. This study utilized spontaneously hypertensive rats (SHRs) as an untr-HT to investigate the impact of far-infrared radiation at specific wavelengths generated by electrified graphene on vascular smooth muscle and blood pressure. After 7 weeks, the blood pressure of the untr-HT group rats decreased significantly with a notable reduction in the number of vascular wall cells and the thickness of the vascular wall, as well as a decreased ratio of vessel wall thickness to lumen diameter. Additionally, blood flow perfusion significantly increased, and the expression of F-actin in vascular smooth muscle myosin decreased significantly. Serum levels of angiotensin II (Ang-II) and endothelin 1 (ET-1) were significantly reduced, while nitric oxide synthase (eNOS) expression increased significantly. At the protein level, eNOS expression decreased significantly, while α-SMA expression increased significantly in aortic tissue. At the gene level, expressions of eNOS and α-SMA in aortic tissue significantly increased. Furthermore, the content of nitric oxide (NO) in the SHR's aortic tissue increased significantly. These findings confirm that graphene far-infrared radiation enhances microcirculation, regulates cytokines affecting vascular smooth muscle contraction, and modifies vascular morphology and smooth muscle phenotype, offering relief for primary hypertension.


Sujet(s)
Pression sanguine , Graphite , Hypertension artérielle , Rayons infrarouges , Muscles lisses vasculaires , Rats de lignée SHR , Animaux , Rats , Pression sanguine/effets des radiations , Mâle , Muscles lisses vasculaires/métabolisme , Graphite/composition chimique , Hypertension artérielle/métabolisme , Nitric oxide synthase type III/métabolisme , Nitric oxide synthase type III/génétique , Angiotensine-II/métabolisme , Angiotensine-II/sang , Endothéline-1/métabolisme , Endothéline-1/génétique , Endothéline-1/sang , Monoxyde d'azote/métabolisme
8.
Eur J Pharmacol ; 977: 176758, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38901528

RÉSUMÉ

Vinclozolin (VCZ) is a common dicarboximide fungicide used to protect crops from diseases. It is also an endocrine disruptor, and its effects on various organs have been described but its influence on vasculature has not yet been addressed. This study focuses on the potential mechanism of VCZ-induced vascular injury. The effect of VCZ on vascular function in terms of relaxing and contracting response was evaluated in mice aorta. A short exposure to VCZ affected the endothelial but not the smooth muscle component. Specifically, it caused a disruption of the eNOS/NO signaling. In line, a short exposure to VCZ in bovine aortic endothelial cells promoted eNOS uncoupling resulting in a reduction of NO bioavailability and eNOS dimer/monomer ratio, and in turn an increase of nitro-tyrosine levels and ROS formation. Prolonging the exposure to VCZ (3 and 6h) an up-regulation of Nox4, enzyme-generating ROS constitutively expressed in endothelial cells, and an increase in ROS and malondialdehyde content coupled with a reduction in NO levels were found. These events were strictly linked to endoplasmic reticulum stress as demonstrated by the phosphorylation of inositol-requiring transmembrane kinase endoribonuclease 1α (IRE1α), a stress sensor and its reversion by using a selective inhibitor. Collectively, these results demonstrated that VCZ provokes endothelial dysfunction by oxidative stress involving eNOS/Nox4/IRE1α axis. The rapid exposure affected the endothelial function promoting eNOS uncoupling while a post-transcriptional modification, involving Nox4/IRE1α signaling, occurred following prolonged exposure. Thus, exposure to VCZ could contribute to the onset and/or progression of cardiovascular diseases associated with endothelial dysfunction.


Sujet(s)
Perturbateurs endocriniens , Endoribonucleases , Cellules endothéliales , NADPH Oxidase 4 , Nitric oxide synthase type III , Monoxyde d'azote , Oxazoles , Protein-Serine-Threonine Kinases , Transduction du signal , Animaux , Nitric oxide synthase type III/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Bovins , Souris , Perturbateurs endocriniens/toxicité , NADPH Oxidase 4/métabolisme , Oxazoles/pharmacologie , Endoribonucleases/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Monoxyde d'azote/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Mâle , Endothélium vasculaire/effets des médicaments et des substances chimiques , Endothélium vasculaire/métabolisme , Endothélium vasculaire/anatomopathologie , Espèces réactives de l'oxygène/métabolisme , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Aorte/effets des médicaments et des substances chimiques , Aorte/métabolisme , Aorte/anatomopathologie
9.
Microcirculation ; 31(5): e12859, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38818977

RÉSUMÉ

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


Sujet(s)
Cellules endothéliales , Gélatine , Microvaisseaux , Nitric oxide synthase type III , Animaux , Cellules endothéliales/métabolisme , Cellules endothéliales/cytologie , Souris , Femelle , Mâle , Microvaisseaux/cytologie , Microvaisseaux/métabolisme , Microvaisseaux/physiologie , Nitric oxide synthase type III/métabolisme , Cellules cultivées , Fibronectines/métabolisme , Fibronectines/pharmacologie , Gels , Antigènes CD/métabolisme , Cadhérines/métabolisme , Culture de cellules primaires , Endothélium vasculaire/métabolisme , Endothélium vasculaire/cytologie
10.
Molecules ; 29(10)2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38792081

RÉSUMÉ

Prolylcarboxypeptidase (PRCP, PCP, Lysosomal Pro-X-carboxypeptidase, Angiotensinase C) controls angiotensin- and kinin-induced cell signaling. Elevation of PRCP appears to be activated in chronic inflammatory diseases [cardiovascular disease (CVD), diabetes] in proportion to severity. Vascular endothelial cell senescence and mitochondrial dysfunction have consistently been shown in models of CVD in aging. Cellular senescence, a driver of age-related dysfunction, can differentially alter the expression of lysosomal enzymes due to lysosomal membrane permeability. There is a lack of data demonstrating the effect of age-related dysfunction on the expression and function of PRCP. To explore the changes in PRCP, the PRCP-dependent prekallikrein (PK) pathway was characterized in early- and late-passage human pulmonary artery endothelial cells (HPAECs). Detailed kinetic analysis of cells treated with high molecular weight kininogen (HK), a precursor of bradykinin (BK), and PK revealed a mechanism by which senescent HPAECs activate the generation of kallikrein upon the assembly of the HK-PK complex on HPAECs in parallel with an upregulation of PRCP and endothelial nitric oxide (NO) synthase (eNOS) and NO formation. The NO production and expression of both PRCP and eNOS increased in early-passage HPAECs and decreased in late-passage HPAECs. Low activity of PRCP in late-passage HPAECs was associated with rapid decreased telomerase reverse transcriptase mRNA levels. We also found that, with an increase in the passage number of HPAECs, reduced PRCP altered the respiration rate. These results indicated that aging dysregulates PRCP protein expression, and further studies will shed light into the complexity of the PRCP-dependent signaling pathway in aging.


Sujet(s)
Marqueurs biologiques , Carboxypeptidases , Vieillissement de la cellule , Cellules endothéliales , Humains , Cellules endothéliales/métabolisme , Marqueurs biologiques/métabolisme , Carboxypeptidases/métabolisme , Carboxypeptidases/génétique , Prékallicréine/métabolisme , Prékallicréine/génétique , Bradykinine/pharmacologie , Bradykinine/métabolisme , Artère pulmonaire/métabolisme , Artère pulmonaire/cytologie , Cellules cultivées , Kininogène de haut poids moléculaire/métabolisme , Transduction du signal , Nitric oxide synthase type III/métabolisme , Nitric oxide synthase type III/génétique , Kallicréines/métabolisme , Kallicréines/génétique
11.
Clin Transl Sci ; 17(5): e13816, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38747311

RÉSUMÉ

Hypertensive patients with a higher proportion of genetic West African ancestry (%GWAA) have better blood pressure (BP) response to thiazide diuretics (TDs) and worse response to ß-blockers (BBs) than those with lower %GWAA, associated with their lower plasma renin activity (PRA). TDs and BBs are suggested to reduce BP in the long term through vasodilation via incompletely understood mechanisms. This study aimed at identifying pathways underlying ancestral differences in PRA, which might reflect pathways underlying BP-lowering mechanisms of TDs and BBs. Among hypertensive participants enrolled in the Pharmacogenomics Evaluation of Antihypertensive Responses (PEAR) and PEAR-2 trials, we previously identified 8 metabolites associated with baseline PRA and 4 metabolic clusters (including 39 metabolites) that are different between those with GWAA <45% versus ≥45%. In the current study, using Ingenuity Pathway Analysis (IPA), we integrated these signals. Three overlapping metabolic signals within three significantly enriched pathways were identified as associated with both PRA and %GWAA: ceramide signaling, sphingosine 1- phosphate signaling, and endothelial nitric oxide synthase signaling. Literature indicates that the identified pathways are involved in the regulation of the Rho kinase cascade, production of the vasoactive agents nitric oxide, prostacyclin, thromboxane A2, and endothelin 1; the pathways proposed to underlie TD- and BB-induced vasodilatation. These findings may improve our understanding of the BP-lowering mechanisms of TDs and BBs. This might provide a possible step forward in personalizing antihypertensive therapy by identifying patients expected to have robust BP-lowering effects from these drugs.


Sujet(s)
Antagonistes bêta-adrénergiques , Pression sanguine , Hypertension artérielle , Métabolomique , Inhibiteurs du symport chlorure sodium , Humains , Mâle , Femelle , Inhibiteurs du symport chlorure sodium/usage thérapeutique , Hypertension artérielle/traitement médicamenteux , Hypertension artérielle/physiopathologie , Pression sanguine/effets des médicaments et des substances chimiques , Adulte d'âge moyen , Antagonistes bêta-adrénergiques/usage thérapeutique , Antagonistes bêta-adrénergiques/pharmacologie , Rénine/sang , Sujet âgé , Nitric oxide synthase type III/métabolisme , Nitric oxide synthase type III/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Adulte
12.
Pregnancy Hypertens ; 36: 101130, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38805888

RÉSUMÉ

OBJECTIVES: Maternal endothelial dysfunction in pregnancy hypertension is related to impairment of nitric oxide (NO) formation. However, NO levels and hemodynamic repercussions on the female offspring remain unclear. Therefore, this study hypothesized that maternal pregnancy hypertension reduces circulating NO metabolites and increases arterial blood pressure in first-generation offspring female rats. STUDY DESIGN: Descendant female rats were distributed in four groups as follows: virgin offspring of normotensive (VN) and hypertensive (VH) mothers and pregnant offspring of normotensive (PN) and hypertensive (PH) mothers. Hemodynamic and biochemical analyses were performed. MAIN OUTCOME MEASURES: The systolic (SBP) and diastolic (DBP) blood pressure, heart rate (HR), and body weight were measured. NO metabolites in plasma, NO formation in human umbilical vein endothelial cells (HUVECs) incubated with plasma, and endothelial NO synthase (eNOS) expression in aortas were determined. RESULTS: Increased SBP, DBP, and reduced HR were found on the 60 days of life in the VH group, whereas the PH group showed increased SBP and HR on pregnancy day 7. All groups showed no differences in body weight gain and eNOS expression. Plasma levels of NO metabolites were increased in the PN compared to the other groups. Increases in the NO formation were greater in HUVECs incubated with plasma from VN and PN groups compared to the VH and PH groups. CONCLUSIONS: Female virgin and pregnant first-generation offspring rats from hypertensive pregnant mothers may have negative cardiovascular repercussions featured by increases in SBP, and possibly impaired NO formation is involved.


Sujet(s)
Monoxyde d'azote , Animaux , Femelle , Grossesse , Monoxyde d'azote/métabolisme , Monoxyde d'azote/sang , Rats , Humains , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Nitric oxide synthase type III/métabolisme , Hypertension artérielle gravidique/physiopathologie , Hypertension artérielle gravidique/métabolisme , Hypertension artérielle gravidique/sang , Effets différés de l'exposition prénatale à des facteurs de risque , Pression artérielle , Modèles animaux de maladie humaine , Pression sanguine/physiologie , Rat Wistar , Rythme cardiaque
13.
Food Chem Toxicol ; 189: 114763, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38797315

RÉSUMÉ

Monosodium glutamate (MSG) administration has been shown to pronounce hypertension and oxidative status with increased renal blood flow (RBF), however, the precise mechanisms of action have never been demonstrated. This study aimed to investigate the MSG action by studying the alteration in renal architecture and specific protein expression in 2-kidney-1-clip hypertensive comparing to sham operative normotensive rats. The administered doses of MSG were 80, 160, or 320 mg/kg BW daily for 8 weeks. Using routine chemical staining, the congestion of glomerular capillaries, a lesser renal corpuscles and glomeruli size, a widen Bowman capsule's space, an increase in mesangial cell proliferation and mesangial matrix, renal interstitial fibrosis, focal cloudy swelling of renal tubular epithelial cells were observed. Immunological study revealed an increase in the expression of N-methyl-D-aspartate receptor (NMDA-R) and endothelial nitric oxide synthase (eNOS) but a decrease in neuronal NOS (nNOS). It is suggested that MSG may upregulate the NMDA-R levels which responsible for the oxidative stress, glomerular injury, and renal interstitial fibrosis. The NMDA-R may also stimulate eNOS overexpression which resulted in renal microvascular dilatation, a raise in RBF and GFR, and natriuresis and diuresis promotion. Long-term exposure of MSG may trigger adaptation of tubuloglomerular feedback through nNOS downregulation.


Sujet(s)
Hypertension artérielle , Rein , Nitric oxide synthase type III , Nitric oxide synthase type I , Récepteurs du N-méthyl-D-aspartate , Glutamate de sodium , Animaux , Nitric oxide synthase type III/métabolisme , Glutamate de sodium/toxicité , Récepteurs du N-méthyl-D-aspartate/métabolisme , Récepteurs du N-méthyl-D-aspartate/génétique , Nitric oxide synthase type I/métabolisme , Mâle , Rats , Rein/effets des médicaments et des substances chimiques , Rein/métabolisme , Rein/anatomopathologie , Hypertension artérielle/induit chimiquement , Hypertension artérielle/physiopathologie , Hypertension artérielle/métabolisme , Rat Wistar
14.
Free Radic Biol Med ; 221: 89-97, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-38735541

RÉSUMÉ

The complex interplay between hydrogen peroxide (H2O2) and nitric oxide (NO) in endothelial cells presents challenges due to technical limitations in simultaneous measurement, hindering the elucidation of their direct relationship. Previous studies have yielded conflicting findings regarding the impact of H2O2 on NO production. To address this problem, we employed genetically encoded biosensors, HyPer7 for H2O2 and geNOps for NO, allowing simultaneous imaging in single endothelial cells. Optimization strategies were implemented to enhance biosensor performance, including camera binning, temperature regulation, and environmental adjustments to mimic physiological normoxia. Our results demonstrate that under ambient oxygen conditions, H2O2 exhibited no significant influence on NO production. Subsequent exploration under physiological normoxia (5 kPa O2) revealed distinct oxidative stress levels characterized by reduced basal HyPer7 signals, enhanced H2O2 scavenging kinetics, and altered responses to pharmacological treatment. Investigation of the relationship between H2O2 and NO under varying oxygen conditions revealed a lack of NO response to H2O2 under hyperoxia (18 kPa O2) but a modest NO response under physiological normoxia (5 kPa O2). Importantly, the NO response was attenuated by l-NAME, suggesting activation of eNOS by endogenous H2O2 generation upon auranofin treatment. Our study highlights the intricate interplay between H2O2 and NO within the endothelial EA.hy926 cell line, emphasizing the necessity for additional research within physiological contexts due to differential response observed under physiological normoxia (5 kPa O2). This further investigation is essential for a comprehensive understanding of the H2O2 and NO signaling considering the physiological effects of ambient O2 levels involved.


Sujet(s)
Techniques de biocapteur , Cellules endothéliales , Peroxyde d'hydrogène , Nitric oxide synthase type III , Monoxyde d'azote , Stress oxydatif , Oxygène , Peroxyde d'hydrogène/métabolisme , Monoxyde d'azote/métabolisme , Humains , Oxygène/métabolisme , Cellules endothéliales/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , Nitric oxide synthase type III/métabolisme , Nitric oxide synthase type III/génétique , Techniques de biocapteur/méthodes , L-NAME/pharmacologie
15.
Sci Rep ; 14(1): 11444, 2024 05 20.
Article de Anglais | MEDLINE | ID: mdl-38769383

RÉSUMÉ

Neonatal sepsis is a major cause of childhood mortality. Limited diagnostic tools and mechanistic insights have hampered our abilities to develop prophylactic or therapeutic interventions. Biomarkers in human neonatal sepsis have been repeatedly identified as associated with dysregulation of angiopoietin signaling and altered arachidonic acid metabolism. We here provide the mechanistic evidence in support of the relevance for these observations. Angiopoetin-1 (Ang-1), which promotes vascular integrity, was decreased in blood plasma of human and murine septic newborns. In preclinical models, administration of Ang-1 provided prophylactic protection from septic death. Arachidonic acid metabolism appears to be functionally connected to Ang-1 via reactive oxygen species (ROS) with a direct role of nitric oxide (NO). Strengthening this intersection via oral administration of arachidonic acid and/or the NO donor L-arginine provided prophylactic as well as therapeutic protection from septic death while also increasing plasma Ang-1 levels among septic newborns. Our data highlight that targeting angiogenesis-associated pathways with interventions that increase Ang-1 activity directly or indirectly through ROS/eNOS provide promising avenues to prevent and/or treat severe neonatal sepsis.


Sujet(s)
Angiopoïétine-1 , Sepsis néonatal , Monoxyde d'azote , Espèces réactives de l'oxygène , Humains , Animaux , Nouveau-né , Angiopoïétine-1/sang , Angiopoïétine-1/métabolisme , Souris , Espèces réactives de l'oxygène/métabolisme , Monoxyde d'azote/métabolisme , Monoxyde d'azote/sang , Acide arachidonique/métabolisme , Acide arachidonique/sang , Femelle , Mâle , Arginine/sang , Arginine/métabolisme , Transduction du signal , Nitric oxide synthase type III/métabolisme , Néovascularisation pathologique/métabolisme , Marqueurs biologiques/sang , Modèles animaux de maladie humaine , Animaux nouveau-nés ,
16.
Sci Rep ; 14(1): 11240, 2024 05 16.
Article de Anglais | MEDLINE | ID: mdl-38755191

RÉSUMÉ

Nao-an Dropping Pill (NADP) is a Chinese patent medicine which commonly used in clinic for ischemic stroke (IS). However, the material basis and mechanism of its prevention or treatment of IS are unclear, then we carried out this study. 52 incoming blood components were resolved by UHPLC-MS/MS from rat serum, including 45 prototype components. The potential active prototype components hydroxysafflor yellow A, ginsenoside F1, quercetin, ferulic acid and caffeic acid screened by network pharmacology showed strongly binding ability with PIK3CA, AKT1, NOS3, NFE2L2 and HMOX1 by molecular docking. In vitro oxygen-glucose deprivation/reperfusion (OGD/R) experimental results showed that NADP protected HA1800 cells from OGD/R-induced apoptosis by affecting the release of LDH, production of NO, and content of SOD and MDA. Meanwhile, NADP could improve behavioral of middle cerebral artery occlusion/reperfusion (MCAO/R) rats, reduce ischemic area of cerebral cortex, decrease brain water and glutamate (Glu) content, and improve oxidative stress response. Immunohistochemical results showed that NADP significantly regulated the expression of PI3K, Akt, p-Akt, eNOS, p-eNOS, Nrf2 and HO-1 in cerebral ischemic tissues. The results suggested that NADP protects brain tissues and ameliorates oxidative stress damage to brain tissues from IS by regulating PI3K/Akt/eNOS and Nrf2/HO-1 signaling pathways.


Sujet(s)
Accident vasculaire cérébral ischémique , Facteur-2 apparenté à NF-E2 , Nitric oxide synthase type III , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Animaux , Facteur-2 apparenté à NF-E2/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Accident vasculaire cérébral ischémique/traitement médicamenteux , Accident vasculaire cérébral ischémique/métabolisme , Accident vasculaire cérébral ischémique/prévention et contrôle , Rats , Phosphatidylinositol 3-kinases/métabolisme , Nitric oxide synthase type III/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Mâle , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/composition chimique , Médicaments issus de plantes chinoises/usage thérapeutique , Rat Sprague-Dawley , Stress oxydatif/effets des médicaments et des substances chimiques , Heme oxygenase-1/métabolisme , Heme oxygenase (decyclizing)/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Humains , Simulation de docking moléculaire
17.
Food Funct ; 15(10): 5485-5495, 2024 May 20.
Article de Anglais | MEDLINE | ID: mdl-38690748

RÉSUMÉ

Ginsenoside Rk1, one kind of ginsenoside, is a minor ginsenoside found in Panax ginseng and used as traditional Chinese medicine for centuries. It exhibits anti-tumor and anti-aggregation effects. However, little research has been done on its effect on endothelial function. This study investigated whether ginsenoside Rk1 improved endothelial dysfunction in diabetes and the underlying mechanisms in vivo and in vitro. Male C57BL/6 mice were fed with a 12 week high-fat diet (60% kcal % fat), whereas treatment groups were orally administered with ginsenoside Rk1 (10 and 20 mg per kg per day) in the last 4 weeks. Aortas isolated from C57BL/6 mice were induced by high glucose (HG; 30 mM) and co-treated with or without ginsenoside Rk1 (1 and 10 µM) for 48 h ex vivo. Moreover, primary rat aortic endothelial cells (RAECs) were cultured and stimulated by HG (44 mM) to mimic hyperglycemia, with or without the co-treatment of ginsenoside Rk1 (10 µM) for 48 h. Endothelium-dependent relaxations of mouse aortas were damaged with elevated oxidative stress and downregulation of three isoforms of peroxisome proliferator-activated receptors (PPARs), PPAR-α, PPAR-ß/δ, and PPAR-γ, as well as endothelial nitric oxide synthase (eNOS) phosphorylation due to HG or high-fat diet stimulation, which also existed in RAECs. However, after the treatment with ginsenoside Rk1, these impairments were all ameliorated significantly. Moreover, the vaso-protective and anti-oxidative effects of ginsenoside Rk1 were abolished by PPAR antagonists (GSK0660, GW9662 or GW6471). In conclusion, this study reveals that ginsenoside Rk1 ameliorates endothelial dysfunction and suppresses oxidative stress in diabetic vasculature through activating the PPAR/eNOS pathway.


Sujet(s)
Endothélium vasculaire , Ginsénosides , Souris de lignée C57BL , Récepteurs activés par les proliférateurs de peroxysomes , Ginsénosides/pharmacologie , Animaux , Mâle , Souris , Rats , Récepteurs activés par les proliférateurs de peroxysomes/métabolisme , Endothélium vasculaire/effets des médicaments et des substances chimiques , Endothélium vasculaire/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Aorte/effets des médicaments et des substances chimiques , Aorte/métabolisme , Nitric oxide synthase type III/métabolisme , Panax/composition chimique , Alimentation riche en graisse
18.
Cardiovasc Diabetol ; 23(1): 150, 2024 May 03.
Article de Anglais | MEDLINE | ID: mdl-38702777

RÉSUMÉ

BACKGROUND: Vasculopathy is the most common complication of diabetes. Endothelial cells located in the innermost layer of blood vessels are constantly affected by blood flow or vascular components; thus, their mechanosensitivity plays an important role in mediating vascular regulation. Endothelial damage, one of the main causes of hyperglycemic vascular complications, has been extensively studied. However, the role of mechanosensitive signaling in hyperglycemic endothelial damage remains unclear. METHODS: Vascular endothelial-specific Piezo1 knockout mice were generated to investigate the effects of Piezo1 on Streptozotocin-induced hyperglycemia and vascular endothelial injury. In vitro activation or knockdown of Piezo1 was performed to evaluate the effects on the proliferation, migration, and tubular function of human umbilical vein endothelial cells in high glucose. Reactive oxygen species production, mitochondrial membrane potential alternations, and oxidative stress-related products were used to assess the extent of oxidative stress damage caused by Piezo1 activation. RESULTS: Our study found that in VECreERT2;Piezo1flox/flox mice with Piezo1 conditional knockout in vascular endothelial cells, Piezo1 deficiency alleviated streptozotocin-induced hyperglycemia with reduced apoptosis and abscission of thoracic aortic endothelial cells, and decreased the inflammatory response of aortic tissue caused by high glucose. Moreover, the knockout of Piezo1 showed a thinner thoracic aortic wall, reduced tunica media damage, and increased endothelial nitric oxide synthase expression in transgenic mice, indicating the relief of endothelial damage caused by hyperglycemia. We also showed that Piezo1 activation aggravated oxidative stress injury and resulted in severe dysfunction through the Ca2+-induced CaMKII-Nrf2 axis in human umbilical vein endothelial cells. In Piezo1 conditional knockout mice, Piezo1 deficiency partially restored superoxide dismutase activity and reduced malondialdehyde content in the thoracic aorta. Mechanistically, Piezo1 deficiency decreased CaMKII phosphorylation and restored the expression of Nrf2 and its downstream molecules HO-1 and NQO1. CONCLUSION: In summary, our study revealed that Piezo1 is involved in high glucose-induced oxidative stress injury and aggravated endothelial dysfunction, which have great significance for alleviating endothelial damage caused by hyperglycemia.


Sujet(s)
Glycémie , Diabète expérimental , Cellules endothéliales de la veine ombilicale humaine , Canaux ioniques , Souris knockout , Nitric oxide synthase type III , Stress oxydatif , Animaux , Humains , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Cellules endothéliales de la veine ombilicale humaine/anatomopathologie , Diabète expérimental/métabolisme , Canaux ioniques/métabolisme , Canaux ioniques/génétique , Glycémie/métabolisme , Nitric oxide synthase type III/métabolisme , Mécanotransduction cellulaire , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Facteur-2 apparenté à NF-E2/déficit , Cellules cultivées , Prolifération cellulaire , Apoptose , Mâle , Angiopathies diabétiques/métabolisme , Angiopathies diabétiques/physiopathologie , Angiopathies diabétiques/anatomopathologie , Angiopathies diabétiques/génétique , Angiopathies diabétiques/étiologie , Mouvement cellulaire , Souris de lignée C57BL , Espèces réactives de l'oxygène/métabolisme , Aorte thoracique/métabolisme , Aorte thoracique/anatomopathologie , Aorte thoracique/physiopathologie , Souris , Streptozocine , Endothélium vasculaire/métabolisme , Endothélium vasculaire/physiopathologie , Endothélium vasculaire/anatomopathologie , Calcium-Calmodulin-Dependent Protein Kinase Type 2/métabolisme , Calcium-Calmodulin-Dependent Protein Kinase Type 2/génétique
19.
Front Biosci (Landmark Ed) ; 29(5): 190, 2024 May 16.
Article de Anglais | MEDLINE | ID: mdl-38812321

RÉSUMÉ

Nitric oxide synthases (NOS) are essential regulators of vascular function, and their role in ocular blood vessels is of paramount importance for maintaining ocular homeostasis. Three isoforms of NOS-endothelial (eNOS), neuronal (nNOS), and inducible (iNOS)-contribute to nitric oxide production in ocular tissues, exerting multifaceted effects on vascular tone, blood flow, and overall ocular homeostasis. Endothelial NOS, primarily located in endothelial cells, is pivotal for mediating vasodilation and regulating blood flow. Neuronal NOS, abundantly found in nerve terminals, contributes to neurotransmitter release and vascular tone modulation in the ocular microvasculature. Inducible NOS, expressed under inflammatory conditions, plays a role in response to pathological stimuli. Understanding the distinctive contributions of these NOS isoforms in retinal blood vessels is vital to unravel the mechanisms underlying various ocular diseases, such diabetic retinopathy. This article delves into the unique contributions of NOS isoforms within the complex vascular network of the retina, elucidating their significance as potential therapeutic targets for addressing pathological conditions.


Sujet(s)
Nitric oxide synthase , Vaisseaux rétiniens , Humains , Vaisseaux rétiniens/métabolisme , Vaisseaux rétiniens/physiopathologie , Animaux , Nitric oxide synthase/métabolisme , Nitric oxide synthase type III/métabolisme , Nitric oxide synthase type I/métabolisme , Monoxyde d'azote/métabolisme , Rétinopathie diabétique/physiopathologie , Rétinopathie diabétique/enzymologie , Rétinopathie diabétique/métabolisme , Nitric oxide synthase type II/métabolisme
20.
Clin Sci (Lond) ; 138(10): 599-614, 2024 May 22.
Article de Anglais | MEDLINE | ID: mdl-38739452

RÉSUMÉ

AIM: Acute kidney injury (AKI) increases the risk for progressive chronic kidney disease (CKD). MicroRNA (miR)-486-5p protects against kidney ischemia-reperfusion (IR) injury in mice, although its long-term effects on the vasculature and development of CKD are unknown. We studied whether miR-486-5p would prevent the AKI to CKD transition in rat, and affect vascular function. METHODS: Adult male rats were subjected to bilateral kidney IR followed by i.v. injection of liposomal-packaged miR-486-5p (0.5 mg/kg). Kidney function and histologic injury were assessed after 24 h and 10 weeks. Kidney endothelial protein levels were measured by immunoblot and immunofluorescence, and mesenteric artery reactivity was determined by wire myography. RESULTS: In rats with IR, miR-486-5p blocked kidney endothelial cell increases in intercellular adhesion molecule-1 (ICAM-1), reduced neutrophil infiltration and histologic injury, and normalized plasma creatinine (P<0.001). However, miR-486-5p attenuated IR-induced kidney endothelial nitric oxide synthase (eNOS) expression (P<0.05). At 10 weeks, kidneys from rats with IR alone had decreased peritubular capillary density and increased interstitial collagen deposition (P<0.0001), and mesenteric arteries showed impaired endothelium-dependent vasorelaxation (P<0.001). These changes were inhibited by miR-486-5p. Delayed miR-486-5p administration (96 h, 3 weeks after IR) had no impact on kidney fibrosis, capillary density, or endothelial function. CONCLUSION: In rats, administration of miR-486-5p early after kidney IR prevents injury, and protects against CKD development and systemic endothelial dysfunction. These protective effects are associated with inhibition of endothelial ICAM-1 and occur despite reduction in eNOS. miR-486-5p holds promise for the prevention of ischemic AKI and its complications.


Sujet(s)
Atteinte rénale aigüe , Molécule-1 d'adhérence intercellulaire , Rein , microARN , Rat Sprague-Dawley , Insuffisance rénale chronique , Lésion d'ischémie-reperfusion , Animaux , microARN/métabolisme , microARN/génétique , Mâle , Atteinte rénale aigüe/prévention et contrôle , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/génétique , Atteinte rénale aigüe/anatomopathologie , Insuffisance rénale chronique/prévention et contrôle , Insuffisance rénale chronique/métabolisme , Insuffisance rénale chronique/génétique , Insuffisance rénale chronique/anatomopathologie , Rein/anatomopathologie , Rein/vascularisation , Rein/métabolisme , Lésion d'ischémie-reperfusion/prévention et contrôle , Lésion d'ischémie-reperfusion/métabolisme , Molécule-1 d'adhérence intercellulaire/métabolisme , Molécule-1 d'adhérence intercellulaire/génétique , Nitric oxide synthase type III/métabolisme , Rats , Modèles animaux de maladie humaine , Évolution de la maladie , Cellules endothéliales/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...