Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 5.964
Filtrer
1.
Sci Rep ; 14(1): 13862, 2024 06 15.
Article de Anglais | MEDLINE | ID: mdl-38879688

RÉSUMÉ

Acute kidney injury (AKI) is a systemic disease that affects energy metabolism in various remote organs in murine models of ischemic AKI. However, AKI-mediated effects in the liver have not been comprehensively assessed. After inducing ischemic AKI in 8-10-week-old, male C57BL/6 mice, mass spectrometry metabolomics revealed that the liver had the most distinct phenotype 24 h after AKI versus 4 h and 7 days. Follow up studies with in vivo [13C6]-glucose tracing on liver and kidney 24 h after AKI revealed 4 major findings: (1) increased flux through glycolysis and the tricarboxylic (TCA) cycle in both kidney and liver; (2) depleted hepatic glutathione levels and its intermediates despite unchanged level of reactive oxygen species, suggesting glutathione consumption exceeds production due to systemic oxidative stress after AKI; (3) hepatic ATP depletion despite unchanged rate of mitochondrial respiration, suggesting increased ATP consumption relative to production; (4) increased hepatic and renal urea cycle intermediates suggesting hypercatabolism and upregulation of the urea cycle independent of impaired renal clearance of nitrogenous waste. Taken together, this is the first study to describe the hepatic metabolome after ischemic AKI in a murine model and demonstrates that there is significant liver-kidney crosstalk after AKI.


Sujet(s)
Atteinte rénale aigüe , Métabolisme énergétique , Glutathion , Rein , Foie , Souris de lignée C57BL , Animaux , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/étiologie , Foie/métabolisme , Glutathion/métabolisme , Rein/métabolisme , Mâle , Souris , Ischémie/métabolisme , Métabolomique/méthodes , Modèles animaux de maladie humaine , Stress oxydatif , Glycolyse , Métabolome
2.
Sci Rep ; 14(1): 13732, 2024 06 14.
Article de Anglais | MEDLINE | ID: mdl-38877069

RÉSUMÉ

Intestinal preservation for transplantation is accompanied by hypoperfusion with long periods of ischemia with total blood cessation and absolute withdrawal of oxygen leading to structural damage. The application of intraluminal oxygen has been successfully tested in small-animal series during storage and transport of the organ but have been so far clinically unrelatable. In this study, we tested whether a simple and clinically approachable method of intraluminal oxygen application could prevent ischemic damage in a large animal model, during warm ischemia time. We utilised a local no-flow ischemia model of the small intestine in pigs. A low-flow and high-pressure intraluminal oxygen deliverance system was applied in 6 pigs and 6 pigs served as a control group. Mucosal histopathology, hypoxia and barrier markers were evaluated after two hours of no-flow conditions, in both treatment and sham groups, and in healthy tissue. Macro- and microscopically, the luminal oxygen delivered treatment group showed preserved small bowel's appearance, viability, and mucosal integrity. A gradual deterioration of histopathology and barrier markers and increase in hypoxia-inducible factor 1-α expression towards the sites most distant from the oxygen application was observed. Intraluminal low-flow, high oxygen delivery can preserve the intestinal mucosa during total ischemia of the small intestine. This finding can be incorporated in methods to overcome small bowel ischemia and improve intestinal preservation for transplantation.


Sujet(s)
Muqueuse intestinale , Intestin grêle , Ischémie , Oxygène , Animaux , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Muqueuse intestinale/vascularisation , Intestin grêle/métabolisme , Intestin grêle/vascularisation , Intestin grêle/anatomopathologie , Oxygène/métabolisme , Suidae , Ischémie/métabolisme , Ischémie/anatomopathologie , Ischémie/thérapie , Modèles animaux de maladie humaine , Conservation d'organe/méthodes , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme
3.
Sci Transl Med ; 16(752): eadf0555, 2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38896604

RÉSUMÉ

Despite decades of effort aimed at developing clinically effective cell therapies, including mixed population mononuclear cells, to revascularize the ischemic limb, there remains a paucity of patient-based studies that inform the function and fate of candidate cell types. In this study, we showed that circulating proangiogenic/arteriogenic monocytes (PAMs) expressing the FcγIIIA receptor CD16 were elevated in patients with chronic limb-threatening ischemia (CLTI), and these amounts decreased after revascularization. Unlike CD16-negative monocytes, PAMs showed large vessel remodeling properties in vitro when cultured with endothelial cells and smooth muscle cells and promoted salvage of the ischemic limb in vivo in a mouse model of hindlimb ischemia. PAMs showed a propensity to migrate toward and bind to ischemic muscle and to secrete angiogenic/arteriogenic factors, vascular endothelial growth factor A (VEGF-A) and heparin-binding epidermal growth factor. We instigated a first-in-human single-arm cohort study in which autologous PAMs were injected into the ischemic limbs of five patients with CLTI. Greater than 25% of injected cells were retained in the leg for at least 72 hours, of which greater than 80% were viable, with evidence of enhanced large vessel remodeling in the injected muscle area. In summary, we identified up-regulation of a circulatory PAM subpopulation as an endogenous response to limb ischemia in CLTI and tested a potentially clinically relevant therapeutic strategy.


Sujet(s)
Membre pelvien , Ischémie , Monocytes , Néovascularisation physiologique , Humains , Monocytes/métabolisme , Animaux , Ischémie/anatomopathologie , Ischémie/métabolisme , Ischémie/thérapie , Membre pelvien/vascularisation , Récepteurs du fragment Fc des IgG/métabolisme , Souris , Mâle , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Femelle , Sujet âgé , Adulte d'âge moyen , Mouvement cellulaire , Facteur de croissance de type EGF liant l'héparine/métabolisme
4.
Clin Transl Med ; 14(6): e1725, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38886900

RÉSUMÉ

BACKGROUND: Angiogenesis is critical for forming new blood vessels from antedating vascular vessels. The endothelium is essential for angiogenesis, vascular remodelling and minimisation of functional deficits following ischaemia. The insulin-like growth factor (IGF) family is crucial for angiogenesis. Insulin-like growth factor-binding protein 5 (IGFBP5), a binding protein of the IGF family, may have places in angiogenesis, but the mechanisms are not yet completely understood. We sought to probe whether IGFBP5 is involved in pathological angiogenesis and uncover the molecular mechanisms behind it. METHODS AND RESULTS: IGFBP5 expression was elevated in the vascular endothelium of gastrocnemius muscle from critical limb ischaemia patients and hindlimb ischaemic (HLI) mice and hypoxic human umbilical vein endothelial cells (HUVECs). In vivo, loss of endothelial IGFBP5 (IGFBP5EKO) facilitated the recovery of blood vessel function and limb necrosis in HLI mice. Moreover, skin damage healing and aortic ring sprouting were faster in IGFBP5EKO mice than in control mice. In vitro, the genetic inhibition of IGFBP5 in HUVECs significantly promoted tube formation, cell proliferation and migration by mediating the phosphorylation of IGF1R, Erk1/2 and Akt. Intriguingly, pharmacological treatment of HUVECs with recombinant human IGFBP5 ensued a contrasting effect on angiogenesis by inhibiting the IGF1 or IGF2 function. Genetic inhibition of IGFBP5 promoted cellular oxygen consumption and extracellular acidification rates via IGF1R-mediated glycolytic adenosine triphosphate (ATP) metabolism. Mechanistically, IGFBP5 exerted its role via E3 ubiquitin ligase Von Hippel-Lindau (VHL)-regulated HIF1α stability. Furthermore, the knockdown of the endothelial IGF1R partially abolished the reformative effect of IGFBP5EKO mice post-HLI. CONCLUSION: Our findings demonstrate that IGFBP5 ablation enhances angiogenesis by promoting ATP metabolism and stabilising HIF1α, implying IGFBP5 is a novel therapeutic target for treating abnormal angiogenesis-related conditions.


Sujet(s)
Membre pelvien , Protéine-5 de liaison aux IGF , Animaux , Protéine-5 de liaison aux IGF/génétique , Protéine-5 de liaison aux IGF/métabolisme , Souris , Membre pelvien/vascularisation , Humains , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Ischémie/métabolisme , Ischémie/génétique , Modèles animaux de maladie humaine , Mâle , Néovascularisation physiologique/génétique ,
5.
J Cell Mol Med ; 28(12): e18489, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38899522

RÉSUMÉ

This study explores the impact of senescence on autocrine C-C motif chemokine ligand 5 (CCL5) in human endothelial progenitor cell (EPCs), addressing the poorly understood decline in number and function of EPCs during ageing. We examined the effects of replication-induced senescence on CCL5/CCL5 receptor (CCR5) signalling and angiogenic activity of EPCs in vitro and in vivo. We also explored microRNAs controlling CCL5 secretion in senescent EPCs, its impact on EPC angiogenic activity, and validated our findings in humans. CCL5 secretion and CCR5 levels in senescent EPCs were reduced, leading to attenuated angiogenic activity. CCL5 enhanced EPC proliferation via the CCR5/AKT/P70S6K axis and increased vascular endothelial growth factor (VEGF) secretion. Up-regulation of miR-409 in senescent EPCs resulted in decreased CCL5 secretion, inhibiting the angiogenic activity, though these negative effects were counteracted by the addition of CCL5 and VEGF. In a mouse hind limb ischemia model, CCL5 improved the angiogenic activity of senescent EPCs. Analysis involving 62 healthy donors revealed a negative association between CCL5 levels, age and Framingham Risk Score. These findings propose CCL5 as a potential biomarker for detection of EPC senescence and cardiovascular risk assessment, suggesting its therapeutic potential for age-related cardiovascular disorders.


Sujet(s)
Vieillissement de la cellule , Chimiokine CCL5 , Progéniteurs endothéliaux , microARN , Néovascularisation physiologique , Chimiokine CCL5/métabolisme , Chimiokine CCL5/génétique , Progéniteurs endothéliaux/métabolisme , Progéniteurs endothéliaux/cytologie , Humains , microARN/génétique , microARN/métabolisme , Animaux , Néovascularisation physiologique/génétique , Souris , Prolifération cellulaire , Mâle , Récepteurs CCR5/métabolisme , Récepteurs CCR5/génétique , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Régulation négative/génétique , Ischémie/métabolisme , Ischémie/anatomopathologie , Ischémie/génétique , Transduction du signal ,
6.
Pharmacol Res ; 205: 107259, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38871237

RÉSUMÉ

The osteopontin-derived peptide FOL-005 stimulates hair growth. Using ligand-receptor glyco-capture technology we identified neuropilin-1 (NRP-1), a known co-receptor for vascular endothelial growth factor (VEGF) receptors, as the most probable receptor for FOL-005 and the more stable analogue FOL-026. X-ray diffraction and microscale thermophoresis analysis revealed that FOL-026 shares binding site with VEGF in the NRP-1 b1-subdomain. Stimulation of human umbilical vein endothelial cells with FOL-026 resulted in phosphorylation of VEGFR-2, ERK1/2 and AKT, increased cell growth and migration, stimulation of endothelial tube formation and inhibition of apoptosis in vitro. FOL-026 also promoted angiogenesis in vivo as assessed by subcutaneous Matrigel plug and hind limb ischemia models. NRP-1 knock-down or treatment of NRP-1 antagonist EG00229 blocked the stimulatory effects of FOL-026 on endothelial cells. Exposure of human coronary artery smooth muscle cells to FOL-026 stimulated cell growth, migration, inhibited apoptosis, and induced VEGF gene expression and VEGFR-2/AKT phosphorylation by an NRP-1-dependent mechanism. RNA sequencing showed that FOL-026 activated pathways involved in tissue repair. These findings identify NRP-1 as the receptor for FOL-026 and show that its biological effects mimic that of growth factors binding to the VEGF receptor family. They also suggest that FOL-026 may have therapeutical potential in conditions that require vascular repair and/or enhanced angiogenesis.


Sujet(s)
Cellules endothéliales de la veine ombilicale humaine , Néovascularisation physiologique , Neuropiline 1 , Ostéopontine , Neuropiline 1/métabolisme , Humains , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Animaux , Néovascularisation physiologique/effets des médicaments et des substances chimiques , Ostéopontine/métabolisme , Ostéopontine/génétique , Mouvement cellulaire/effets des médicaments et des substances chimiques , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/métabolisme , Mâle , Peptides/pharmacologie , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Liaison aux protéines , Ischémie/traitement médicamenteux , Ischémie/métabolisme , Souris ,
7.
Am J Physiol Heart Circ Physiol ; 327(1): H268-H274, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38787380

RÉSUMÉ

Brachial artery flow-mediated dilation (BAFMD) is induced by hyperemic wall shear rate (WSR) following forearm ischemia. In older adults, there appears to be a reduced brachial hyperemic WSR and altered stimulus-response relationship compared with young adults. However, it is unclear if an altered forearm microvascular response to ischemia influences brachial hyperemic WSR in older adults. We determined associations between brachial hyperemic WSR and forearm skeletal muscle oxygen saturation in young and older adults. Healthy young (n = 17, 29 ± 7 yr) and older (n = 32, 65 ± 4 yr) adults participated in the study. BAFMD by a multigate spectral Doppler system and forearm skeletal muscle oxygen saturation by near-infrared spectroscopy were concurrently measured. When compared with the young, older adults showed reduced oxygen extraction kinetics (OE, 0.15 [0.12-0.17] vs. 0.09 [0.05-0.12]%s-1) and magnitude (So2deficit, 3,810 ± 1,420 vs. 2,723 ± 1,240%s) during ischemia, as well as oxygen resaturation kinetics (So2slope, 2.5 ± 0.7 vs. 1.7 ± 0.7%s-1) upon reperfusion (all P < 0.05). When OE in the young and So2slope in older adults were stratified by their median values, young adults with OE above the median had greater hyperemic WSR parameters compared with those below the median (P < 0.05), but So2slope in older adults did not show clear differences in hyperemic WSR parameters between those above/below the median. This study demonstrates that, in addition to a reduced microvascular response to ischemia, there may be a dissociation between microvascular response to ischemia and brachial hyperemic WSR in older adults, which may result in a further impairment of BAFMD in this cohort.NEW & NOTEWORTHY Microvascular response to ischemia and subsequent reperfusion is diminished in older adults compared with the young. Furthermore, there appears to be a dissociation between the microvascular response to ischemia and brachial hyperemic WSR in older adults, which may further disturb the BAFMD process in this cohort. A reduced BAFMD in older adults may be a result of multiple alterations occurring both at macro- and microcirculation.


Sujet(s)
Artère brachiale , Avant-bras , Hyperhémie , Microcirculation , Muscles squelettiques , Débit sanguin régional , Vasodilatation , Humains , Artère brachiale/physiopathologie , Artère brachiale/imagerie diagnostique , Mâle , Femelle , Adulte , Sujet âgé , Hyperhémie/physiopathologie , Hyperhémie/métabolisme , Muscles squelettiques/vascularisation , Muscles squelettiques/métabolisme , Adulte d'âge moyen , Avant-bras/vascularisation , Jeune adulte , Ischémie/physiopathologie , Ischémie/métabolisme , Facteurs âges , Vitesse du flux sanguin , Spectroscopie proche infrarouge , Vieillissement/métabolisme , Vieillissement/physiologie , Consommation d'oxygène , Saturation en oxygène , Microvaisseaux/physiopathologie , Microvaisseaux/métabolisme , Microvaisseaux/imagerie diagnostique
8.
Nature ; 629(8012): 660-668, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38693258

RÉSUMÉ

Ischaemic diseases such as critical limb ischaemia and myocardial infarction affect millions of people worldwide1. Transplanting endothelial cells (ECs) is a promising therapy in vascular medicine, but engrafting ECs typically necessitates co-transplanting perivascular supporting cells such as mesenchymal stromal cells (MSCs), which makes clinical implementation complicated2,3. The mechanisms that enable MSCs to facilitate EC engraftment remain elusive. Here we show that, under cellular stress, MSCs transfer mitochondria to ECs through tunnelling nanotubes, and that blocking this transfer impairs EC engraftment. We devised a strategy to artificially transplant mitochondria, transiently enhancing EC bioenergetics and enabling them to form functional vessels in ischaemic tissues without the support of MSCs. Notably, exogenous mitochondria did not integrate into the endogenous EC mitochondrial pool, but triggered mitophagy after internalization. Transplanted mitochondria co-localized with autophagosomes, and ablation of the PINK1-Parkin pathway negated the enhanced engraftment ability of ECs. Our findings reveal a mechanism that underlies the effects of mitochondrial transfer between mesenchymal and endothelial cells, and offer potential for a new approach for vascular cell therapy.


Sujet(s)
Thérapie cellulaire et tissulaire , Cellules endothéliales , Ischémie , Mitochondries , Mitophagie , Animaux , Humains , Mâle , Souris , Autophagosomes/métabolisme , Cellules endothéliales/cytologie , Cellules endothéliales/métabolisme , Cellules endothéliales/transplantation , Métabolisme énergétique , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Ischémie/métabolisme , Ischémie/thérapie , Cellules souches mésenchymateuses/cytologie , Cellules souches mésenchymateuses/métabolisme , Souris nude , Mitochondries/métabolisme , Mitochondries/transplantation , Protein kinases/déficit , Protein kinases/métabolisme , Ubiquitin-protein ligases/déficit , Ubiquitin-protein ligases/métabolisme , Thérapie cellulaire et tissulaire/méthodes
9.
Biochem Biophys Res Commun ; 716: 150002, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38697011

RÉSUMÉ

Type 2 diabetes mellitus (T2DM) significantly impairs the functionality and number of endothelial progenitor cells (EPCs) and resident endothelial cells, critical for vascular repair and regeneration, exacerbating the risk of vascular complications. GLP-1 receptor agonists, like dulaglutide, have emerged as promising therapeutic agents due to their multifaceted effects, including the enhancement of EPC activity and protection of endothelial cells. This study investigates dulaglutide's effects on peripheral blood levels of CD34+ and CD133+ cells in a mouse model of lower limb ischemia and its protective mechanisms against high-glucose-induced damage in endothelial cells. Results demonstrated that dulaglutide significantly improves blood flow, reduces tissue damage and inflammation in ischemic limbs, and enhances glycemic control. Furthermore, dulaglutide alleviated high-glucose-induced endothelial cell damage, evident from improved tube formation, reduced reactive oxygen species accumulation, and restored endothelial junction integrity. Mechanistically, dulaglutide mitigated mitochondrial fission in endothelial cells under high-glucose conditions, partly through maintaining SIRT1 expression, which is crucial for mitochondrial dynamics. This study reveals the potential of dulaglutide as a therapeutic option for vascular complications in T2DM patients, highlighting its role in improving endothelial function and mitochondrial integrity.


Sujet(s)
Diabète expérimental , Progéniteurs endothéliaux , Peptides glucagon-like , Glucose , Fragments Fc des immunoglobulines , Dynamique mitochondriale , Protéines de fusion recombinantes , Sirtuine-1 , Animaux , Fragments Fc des immunoglobulines/pharmacologie , Peptides glucagon-like/analogues et dérivés , Peptides glucagon-like/pharmacologie , Peptides glucagon-like/usage thérapeutique , Sirtuine-1/métabolisme , Dynamique mitochondriale/effets des médicaments et des substances chimiques , Progéniteurs endothéliaux/effets des médicaments et des substances chimiques , Progéniteurs endothéliaux/métabolisme , Protéines de fusion recombinantes/pharmacologie , Mâle , Souris , Glucose/métabolisme , Diabète expérimental/métabolisme , Diabète expérimental/traitement médicamenteux , Diabète expérimental/anatomopathologie , Souris de lignée C57BL , Diabète de type 2/traitement médicamenteux , Diabète de type 2/métabolisme , Diabète de type 2/anatomopathologie , Hypoglycémiants/pharmacologie , Humains , Ischémie/métabolisme , Ischémie/traitement médicamenteux , Ischémie/anatomopathologie
10.
Biomolecules ; 14(5)2024 May 16.
Article de Anglais | MEDLINE | ID: mdl-38785999

RÉSUMÉ

Recently, the vascular protective effect of anti-diabetic agents has been receiving much attention. Sodium glucose cotransporter 2 (SGLT2) inhibitors had demonstrated reductions in cardiovascular (CV) events. However, the therapeutic effect of dapagliflozin on angiogenesis in peripheral arterial disease was unclear. This study aimed to explore the effect and mechanism of dapagliflozin on angiogenesis after hindlimb ischemia. We first evaluated the effect of dapagliflozin on post-ischemic angiogenesis in the hindlimbs of rats. Laser doppler imaging was used to detect the hindlimb blood perfusion. In addition, we used immunohistochemistry to detect the density of new capillaries after ischemia. The relevant signaling pathways of dapagliflozin affecting post-ischemic angiogenesis were screened through phosphoproteomic detection, and then the mechanism of dapagliflozin affecting post-ischemic angiogenesis was verified at the level of human umbilical vein endothelial cells (HUVECs). After subjection to excision of the left femoral artery, all rats were randomly distributed into two groups: the dapagliflozin group (left femoral artery resection, receiving intragastric feeding with dapagliflozin (1 mg/kg/d), for 21 consecutive days) and the model group, that is, the positive control group (left femoral artery resection, receiving intragastric feeding with citric acid-sodium citrate buffer solution (1 mg/kg/d), for 21 consecutive days). In addition, the control group, that is the negative control group (without left femoral artery resection, receiving intragastric feeding with citric acid-sodium citrate buffer solution (1 mg/kg/d), for 21 consecutive days) was added. At day 21 post-surgery, the dapagliflozin-treatment group had the greatest blood perfusion, accompanied by elevated capillary density. The results showed that dapagliflozin could promote angiogenesis after hindlimb ischemia. Then, the ischemic hindlimb adductor-muscle tissue samples from three rats of model group and dapagliflozin group were taken for phosphoproteomic testing. The results showed that the PI3K-Akt-eNOS signaling pathway was closely related to the effect of dapagliflozin on post-ischemic angiogenesis. Our study intended to verify this mechanism from the perspective of endothelial cells. In vitro, dapagliflozin enhanced the tube formation, migration, and proliferation of HUVECs under ischemic and hypoxic conditions. Additionally, the dapagliflozin administration upregulated the expression of angiogenic factors phosphorylated Akt (p-Akt) and phosphorylated endothelial nitric oxide synthase (p-eNOS), as well as vascular endothelial growth factor A (VEGFA), both in vivo and in vitro. These benefits could be blocked by either phosphoinositide 3-kinase (PI3K) or eNOS inhibitor. dapagliflozin could promote angiogenesis after ischemia. This effect might be achieved by promoting the activation of the PI3K-Akt-eNOS signaling pathway. This study provided a new perspective, new ideas, and a theoretical basis for the treatment of peripheral arterial disease.


Sujet(s)
Composés benzhydryliques , Glucosides , Membre pelvien , Cellules endothéliales de la veine ombilicale humaine , Ischémie , Néovascularisation physiologique , Nitric oxide synthase type III , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Animaux , Glucosides/pharmacologie , Composés benzhydryliques/pharmacologie , Membre pelvien/vascularisation , Nitric oxide synthase type III/métabolisme , Ischémie/traitement médicamenteux , Ischémie/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Rats , Humains , Transduction du signal/effets des médicaments et des substances chimiques , Mâle , Néovascularisation physiologique/effets des médicaments et des substances chimiques , Rat Sprague-Dawley ,
11.
Sci Rep ; 14(1): 11372, 2024 05 18.
Article de Anglais | MEDLINE | ID: mdl-38762650

RÉSUMÉ

The aim of this study was to identify angiogenic microRNAs (miRNAs) that could be used in the treatment of hindlimb ischemic tissues. miRNAs contained in extracellular vesicles (EVs) deriving from the plasma were analyzed in C57BL/6 mice, which have ischemia tolerance, and in BALB/c mice without ischemia tolerance as part of a hindlimb ischemia model; as a result 43 angiogenic miRNA candidates were identified. An aortic ring assay was employed by using femoral arteries isolated from BALC/c mice and EVs containing miRNA; as a result, the angiogenic miRNA candidates were limited to 14. The blood flow recovery was assessed after injecting EVs containing miRNA into BALB/c mice with hindlimb ischemia, and miR-709 was identified as a promising angiogenic miRNA. miR-709-encapsulating EVs were found to increase the expression levels of the fibroblast growth factor 2 (FGF2) mRNA in the thigh tissues of hindlimb ischemia model BALB/c mice. miR-709 was also found to bind to the 3'UTR of glycogen synthase kinase 3 beta (GSK3B) in three places. GSK3B-knockdown human artery-derived endothelial cells were found to express high levels of FGF2, and were characterized by increased cell proliferation. These findings indicate that miR-709 induces an upregulation of FGF2 through the downregulation of GSK3B.


Sujet(s)
Facteur de croissance fibroblastique de type 2 , Glycogen synthase kinase 3 beta , Membre pelvien , Ischémie , Souris de lignée BALB C , microARN , Néovascularisation physiologique , Animaux , Humains , Mâle , Souris , Régions 3' non traduites , Prolifération cellulaire , Modèles animaux de maladie humaine , Régulation négative , Cellules endothéliales/métabolisme , Vésicules extracellulaires/métabolisme , Facteur de croissance fibroblastique de type 2/métabolisme , Facteur de croissance fibroblastique de type 2/génétique , Glycogen synthase kinase 3 beta/métabolisme , Glycogen synthase kinase 3 beta/génétique , Membre pelvien/vascularisation , Ischémie/métabolisme , Ischémie/génétique , Souris de lignée C57BL , microARN/génétique , microARN/métabolisme , Néovascularisation physiologique/génétique , Régulation positive
12.
Am J Physiol Heart Circ Physiol ; 327(1): H118-H130, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38758130

RÉSUMÉ

One of the initiating events in preeclampsia (PE) is placental ischemia. Rodent models of placental ischemia do not present with vascular endothelial dysfunction, a hallmark of PE. We previously demonstrated a role for leptin in endothelial dysfunction in pregnancy in the absence of placental ischemia. We hypothesized that placental ischemia requires hyperleptinemia and endothelial mineralocorticoid receptor (ECMR) expression to induce PE-associated endothelial dysfunction in pregnant mice. We induced placental ischemia via the reduced uterine perfusion pressure (RUPP) procedure in pregnant ECMR-intact (ECMR+/+) and ECMR deletion (ECMR-/-) mice at gestational day (GD) 13. ECMR+/+ RUPP pregnant mice also received concurrent leptin infusion via miniosmotic pump (0.9 mg/kg/day). RUPP increased blood pressure via radiotelemetry and decreased fetal growth in ECMR+/+ pregnant mice. Both increases in blood pressure and reduced fetal growth were abolished in RUPP ECMR-/- mice. Placental ischemia did not decrease endothelial-dependent relaxation to acetylcholine (ACh) but increased phenylephrine (Phe) contraction in mesenteric arteries of pregnant mice, which was ablated by ECMR deletion. Addition of leptin to RUPP mice significantly reduced ACh relaxation in ECMR+/+ pregnant mice, accompanied by an increase in soluble FMS-like tyrosine kinase-1 (sFlt-1)/placental growth factor (PLGF) ratio. In conclusion, our data indicate that high leptin levels drive endothelial dysfunction in PE and that ECMR is required for clinical characteristics of hypertension and fetal growth restriction in placental ischemia PE. Collectively, we show that both ECMR and leptin play a role to mediate PE.NEW & NOTEWORTHY Leptin is a key feature of preeclampsia that initiates vascular endothelial dysfunction in preeclampsia characterized by placental ischemia. Endothelial mineralocorticoid receptor (ECMR) deletion in placental ischemia protects pregnant mice from elevations in blood pressure and fetal growth restriction in pregnancy. Increases in leptin production mediate the key pathological feature of endothelial dysfunction in preeclampsia in rodents. ECMR activation contributes to the increase in blood pressure and fetal growth restriction in preeclampsia.


Sujet(s)
Ischémie , Leptine , Placenta , Pré-éclampsie , Récepteurs des minéralocorticoïdes , Animaux , Grossesse , Femelle , Leptine/métabolisme , Leptine/sang , Placenta/métabolisme , Placenta/vascularisation , Ischémie/physiopathologie , Ischémie/métabolisme , Ischémie/génétique , Récepteurs des minéralocorticoïdes/métabolisme , Récepteurs des minéralocorticoïdes/génétique , Pré-éclampsie/métabolisme , Pré-éclampsie/physiopathologie , Pré-éclampsie/génétique , Souris knockout , Pression sanguine , Souris de lignée C57BL , Souris , Modèles animaux de maladie humaine , Retard de croissance intra-utérin/métabolisme , Retard de croissance intra-utérin/physiopathologie , Retard de croissance intra-utérin/génétique , Endothélium vasculaire/métabolisme , Endothélium vasculaire/physiopathologie , Vasodilatation/effets des médicaments et des substances chimiques
13.
PLoS One ; 19(5): e0303758, 2024.
Article de Anglais | MEDLINE | ID: mdl-38768136

RÉSUMÉ

Nitric oxide (NO) promotes angiogenesis via various mechanisms; however, the effective transmission of NO in ischemic diseases is unclear. Herein, we tested whether NO-releasing nanofibers modulate therapeutic angiogenesis in an animal hindlimb ischemia model. Male wild-type C57BL/6 mice with surgically-induced hindlimb ischemia were treated with NO-releasing 3-methylaminopropyltrimethoxysilane (MAP3)-derived or control (i.e., non-NO-releasing) nanofibers, by applying them to the wound for 20 min, three times every two days. The amount of NO from the nanofiber into tissues was assessed by NO fluorometric assay. The activity of cGMP-dependent protein kinase (PKG) was determined by western blot analysis. Perfusion ratios were measured 2, 4, and 14 days after inducing ischemia using laser doppler imaging. On day 4, Immunohistochemistry (IHC) with F4/80 and gelatin zymography were performed. IHC with CD31 was performed on day 14. To determine the angiogenic potential of NO-releasing nanofibers, aorta-ring explants were treated with MAP3 or control fiber for 20 min, and the sprout lengths were examined after 6 days. As per either LDPI (Laser doppler perfusion image) ratio or CD31 capillary density measurement, angiogenesis in the ischemic hindlimb was improved in the MAP3 nanofiber group; further, the total nitrate/nitrite concentration in the adduct muscle increased. The number of macrophage infiltrations and matrix metalloproteinase-9 (MMP-9) activity decreased. Vasodilator-stimulated phosphoprotein (VASP), one of the major substrates for PKG, increased phosphorylation in the MAP3 group. MAP3 nanofiber or NO donor SNAP (s-nitroso-n-acetyl penicillamine)-treated aortic explants showed enhanced sprouting in an ex vivo aortic ring assay, which was partially abrogated by KT5823, a potent inhibitor of PKG. These findings suggest that the novel NO-releasing nanofiber, MAP3 activates PKG and promotes therapeutic angiogenesis in response to hindlimb ischemia.


Sujet(s)
Cyclic GMP-Dependent Protein Kinases , Membre pelvien , Ischémie , Souris de lignée C57BL , Nanofibres , Néovascularisation physiologique , Monoxyde d'azote , Animaux , Nanofibres/composition chimique , Mâle , Monoxyde d'azote/métabolisme , Ischémie/traitement médicamenteux , Ischémie/métabolisme , Cyclic GMP-Dependent Protein Kinases/métabolisme , Souris , Membre pelvien/vascularisation , Néovascularisation physiologique/effets des médicaments et des substances chimiques , Matrix metalloproteinase 9/métabolisme , Phosphoprotéines/métabolisme , Protéines des microfilaments/métabolisme , Molécules d'adhérence cellulaire
14.
Adv Sci (Weinh) ; 11(24): e2307238, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38639443

RÉSUMÉ

Preventing and treating avascular necrosis at the distal end of the flaps are critical to surgery success, but current treatments are not ideal. A recent study shows that apoptotic bodies (ABs) generated near the site of apoptosis can be taken up and promote cell proliferation. The study reveals that ABs derived from fibroblast-like cells in the subcutaneous connective tissue (FSCT cells) of skin flaps promoted ischaemic flap survival. It is also found that ABs inhibited cell death and oxidative stress and promoted M1-to-M2 polarization in macrophages. Transcriptome sequencing and protein level testing demonstrated that ABs promoted ischaemic flap survival in endothelial cells and macrophages by inhibiting ferroptosis via the KEAP1-Nrf2 axis. Furthermore, microRNA (miR) sequencing data and in vitro and in vivo experiments demonstrated that ABs inhibited KEAP1 by delivering miR-339-5p to exert therapeutic effects. In conclusion, FSCT cell-derived ABs inhibited ferroptosis, promoted the macrophage M1-to-M2 transition via the miR-339-5p/KEAP1/Nrf2 axis and promoted ischaemic flap survival. These results provide a potential therapeutic strategy to promote ischaemic flap survival by administering ABs.


Sujet(s)
Ferroptose , Fibroblastes , Protéine-1 de type kelch associée à ECH , microARN , Facteur-2 apparenté à NF-E2 , Lambeaux chirurgicaux , Animaux , Souris , Protéine-1 de type kelch associée à ECH/métabolisme , Protéine-1 de type kelch associée à ECH/génétique , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Ferroptose/génétique , microARN/génétique , microARN/métabolisme , Fibroblastes/métabolisme , Modèles animaux de maladie humaine , Ischémie/métabolisme , Ischémie/génétique , Mâle , Apoptose/génétique , Tissu conjonctif/métabolisme , Transduction du signal/génétique
15.
Int J Mol Med ; 53(5)2024 05.
Article de Anglais | MEDLINE | ID: mdl-38577949

RÉSUMÉ

Several studies have shown that berberine (BBR) is effective in protecting against myocardial ischemia­reperfusion injury (MI/RI). However, the precise molecular mechanism remains elusive. The present study observed the mechanism and the safeguarding effect of BBR against hypoxia/reoxygenation (H/R) myocardial injury in H9c2 cells. BBR pretreatment significantly improved the decrease of cell viability, P62 protein, Rho Family GTPase 3 (RhoE) protein, ubiquinone subunit B8 protein, ubiquinol­cytochrome c reductase core protein U, the Bcl­2­associated X protein/B­cell lymphoma 2 ratio, glutathione (GSH) and the GSH/glutathione disulphide (GSSG) ratio induced by H/R, while reducing the increase in lactate dehydrogenase, microtubule­associated protein 1 light 3 protein, caspase­3 activity, reactive oxygen species, GSSG and malonaldehyde caused by H/R. Transmission electron microscopy and LysoTracker Red DND­99 staining results showed that BBR pretreatment inhibited H/R­induced excessive autophagy by mediating RhoE. BBR also inhibited mitochondrial permeability transition, maintained the stability of the mitochondrial membrane potential, reduced the apoptotic rate, and increased the level of caspase­3. However, the protective effects of BBR were attenuated by pAD/RhoE­small hairpin RNA, rapamycin (an autophagy activator) and compound C (an AMP­activated protein kinase inhibitor). These new findings suggested that BBR protects the myocardium from MI/RI by inhibiting excessive autophagy, maintaining mitochondrial function, improving the energy supply and redox homeostasis, and attenuating apoptosis through the RhoE/AMP­activated protein kinase pathway.


Sujet(s)
AMP-Activated Protein Kinases , Autophagie , Berbérine , Lésion de reperfusion myocardique , AMP-Activated Protein Kinases/métabolisme , Apoptose , Berbérine/pharmacologie , Caspase-3/métabolisme , Disulfure de glutathion/métabolisme , Ischémie/métabolisme , Lésion de reperfusion myocardique/traitement médicamenteux , Lésion de reperfusion myocardique/prévention et contrôle , Lésion de reperfusion myocardique/étiologie , Myocarde/anatomopathologie , Myocytes cardiaques/métabolisme , Animaux , Rats
16.
Hypertens Res ; 47(6): 1654-1667, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38605142

RÉSUMÉ

Vascular inflammation and endothelial dysfunction contribute to vascular diseases. While neutrophil extracellular traps (NETs) participate in some vascular pathologies, their roles in lower limb ischemia remain poorly defined. This study investigated the functional significance of NETs in vascular inflammation and remodeling associated with limb ischemia. Single-cell RNA sequencing (scRNA-seq) and flow cytometry revealed neutrophil activation and upregulated NETs formation in human limb ischemia, with immunofluorescence confirming IL-1ß-induced release of NETs for vascular inflammation. Endothelial cell activation was examined via scRNA-seq and western blotting, indicating enhanced proliferation, expression of adhesion molecules (VCAM-1, ICAM-1), inflammatory cytokines (IL-1ß, IL-6) and decreased expression of VE-cadherin, that could be mediated by NETs to exacerbate endothelial inflammation. Mechanistically, NETs altered endothelial cell function via increased pSTAT1/STAT1 signaling. Vascular inflammation and subsequent ischemia were alleviated in vivo by NETosis or IL-1ß inhibition in ischemic mice. IL-1ß-NETs induce endothelial activation and inflammation in limb ischemia by stimulating STAT1 signaling. Targeting NETs may thus represent a novel therapeutic strategy for inflammatory vascular diseases associated with limb ischemia. Graphical abstract of NETs regulation of the development of vascular inflammation in lower limb ischemia via pSTAT1/STAT1 signaling pathway.


Sujet(s)
Pièges extracellulaires , Interleukine-1 bêta , Ischémie , Pièges extracellulaires/métabolisme , Interleukine-1 bêta/métabolisme , Ischémie/métabolisme , Ischémie/physiopathologie , Animaux , Humains , Souris , Mâle , Facteur de transcription STAT-1/métabolisme , Endothélium vasculaire/métabolisme , Souris de lignée C57BL , Granulocytes neutrophiles , Transduction du signal , Membre inférieur/vascularisation , Cellules endothéliales/métabolisme
17.
J Am Heart Assoc ; 13(9): e029880, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38639336

RÉSUMÉ

BACKGROUND: Cellular therapies have been investigated to improve blood flow and prevent amputation in peripheral artery disease with limited efficacy in clinical trials. Alginate-encapsulated mesenchymal stromal cells (eMSCs) demonstrated improved retention and survival and promoted vascular generation in murine hind limb ischemia through their secretome, but large animal evaluation is necessary for human applicability. We sought to determine the efficacy of eMSCs for peripheral artery disease-induced limb ischemia through assessment in our durable swine hind limb ischemia model. METHODS AND RESULTS: Autologous bone marrow eMSCs or empty alginate capsules were intramuscularly injected 2 weeks post-hind limb ischemia establishment (N=4/group). Improvements were quantified for 4 weeks through walkway gait analysis, contrast angiography, blood pressures, fluorescent microsphere perfusion, and muscle morphology and histology. Capsules remained intact with mesenchymal stromal cells retained for 4 weeks. Adenosine-induced perfusion deficits and muscle atrophy in ischemic limbs were significantly improved by eMSCs versus empty capsules (mean±SD, 1.07±0.19 versus 0.41±0.16, P=0.002 for perfusion ratios and 2.79±0.12 versus 1.90±0.62 g/kg, P=0.029 for ischemic muscle mass). Force- and temporal-associated walkway parameters normalized (ratio, 0.63±0.35 at week 3 versus 1.02±0.19 preligation; P=0.17), and compensatory footfall patterning was diminished in eMSC-administered swine (12.58±8.46% versus 34.85±15.26%; P=0.043). Delivery of eMSCs was associated with trending benefits in collateralization, local neovascularization, and muscle fibrosis. Hypoxia-cultured porcine mesenchymal stromal cells secreted vascular endothelial growth factor and tissue inhibitor of metalloproteinase 2. CONCLUSIONS: This study demonstrates the promise of the mesenchymal stromal cell secretome at improving peripheral artery disease outcomes and the potential for this novel swine model to serve as a component of the preclinical pipeline for advanced therapies.


Sujet(s)
Alginates , Modèles animaux de maladie humaine , Membre pelvien , Ischémie , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Animaux , Transplantation de cellules souches mésenchymateuses/méthodes , Membre pelvien/vascularisation , Cellules souches mésenchymateuses/métabolisme , Ischémie/physiopathologie , Ischémie/thérapie , Ischémie/métabolisme , Suidae , Néovascularisation physiologique , Maladie artérielle périphérique/thérapie , Maladie artérielle périphérique/physiopathologie , Maladie artérielle périphérique/anatomopathologie , Injections musculaires , Débit sanguin régional , Muscles squelettiques/vascularisation , , Cellules cultivées
18.
Eur J Pharmacol ; 974: 176621, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38679118

RÉSUMÉ

BACKGROUND AND AIM: Necrosis of random-pattern flaps restricts their application in clinical practice. Puerarin has come into focus due to its promising therapeutic effects in ischemic diseases. Here, we employed Puerarin and investigated its role and potential mechanisms in flap survival. EXPERIMENTAL PROCEDURE: The effect of Puerarin on the viability of human umbilical vein endothelial cells (HUVECs) was assessed by CCK-8, EdU staining, migration, and scratch assays. Survival area measurement and laser Doppler blood flow (LDBF) were utilized to assess the viability of ischemic injury flaps. Levels of molecules related to oxidative stress, pyroptosis, autophagy, transcription factor EB (TFEB), and the AMPK-TRPML1-Calcineurin signaling pathway were detected using western blotting, immunofluorescence, dihydroethidium (DHE) staining, RT-qPCR and Elisa. KEY RESULTS: The findings demonstrated that Puerarin enhanced the survivability of ischemic flaps. Autophagy, oxidative stress, and pyroptosis were implicated in the ability of Puerarin in improving flap survival. Increased autophagic flux and augmented tolerance to oxidative stress contribute to Puerarin's suppression of pyroptosis. Additionally, Puerarin modulated the activity of TFEB through the AMPK-TRPML1-Calcineurin signaling pathway, thereby enhancing autophagic flux. CONCLUSIONS AND IMPLICATIONS: Puerarin promoted flap survival from ischemic injury through upregulation of TFEB-mediated autophagy and inhibition of oxidative stress. Our findings offered valuable support for the clinical application of Puerarin in the treatment of ischemic diseases, including random-pattern flaps.


Sujet(s)
Autophagie , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines , Cellules endothéliales de la veine ombilicale humaine , Ischémie , Isoflavones , Pyroptose , Espèces réactives de l'oxygène , Isoflavones/pharmacologie , Isoflavones/usage thérapeutique , Autophagie/effets des médicaments et des substances chimiques , Humains , Pyroptose/effets des médicaments et des substances chimiques , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/métabolisme , Ischémie/traitement médicamenteux , Ischémie/métabolisme , Espèces réactives de l'oxygène/métabolisme , Animaux , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Mâle , Stress oxydatif/effets des médicaments et des substances chimiques , Lambeaux chirurgicaux/vascularisation , Souris , Transduction du signal/effets des médicaments et des substances chimiques , Peau/effets des médicaments et des substances chimiques , Peau/métabolisme , Peau/vascularisation , Peau/anatomopathologie
19.
Acta Cir Bras ; 39: e391724, 2024.
Article de Anglais | MEDLINE | ID: mdl-38629650

RÉSUMÉ

PURPOSE: To investigate inflammation and cell adhesion molecules in the vagina after ovarian ischemia-reperfusion (IR) injury. METHODS: 20 Wistar albino female rats were divided into two groups: control, and IR groups. In IR group, blood flow was restricted for 2 hours for ovarian ischemia. Then, tissues were re-blood 2 hours for reperfusion. Vagina tissues were excised and processed for histopathological analysis. Histopathological and biochemical follow-ups were performed. RESULTS: Both malondialdehyde and myeloperoxidase values were increased in IR group compared to control group. Glutathione content was decreased in IR group compared to control group. Epithelial degeneration, inflammation, dilatation, and nuclear factor-κB (NF-κB) expression were increased in IR group compared to control group. E-cadherin expression was significantly decreased in IR group. In the IR group, E-cadherin showed a positive reaction in adenomas, gland-like cryptic structures, cellular junctions with clustered inflammatory cells. In the IR group, NF-κB expression was increased in basement membrane, inflammatory cells, in blood vessels. CONCLUSIONS: Ovarian ischemia caused degeneration of epithelial cells in the vaginal region and disruptions in the cell junction complex, which leads to activation of E-cadherin and NF-κB signaling pathway and alterations in reproductive and embryonal development in the vaginal region.


Sujet(s)
Cadhérines , Facteur de transcription NF-kappa B , Lésion d'ischémie-reperfusion , Animaux , Femelle , Rats , Cadhérines/métabolisme , Inflammation , Ischémie/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Rat Wistar , Lésion d'ischémie-reperfusion/anatomopathologie , Ovaire/anatomopathologie , Vagin/métabolisme , Vagin/anatomopathologie
20.
Fluids Barriers CNS ; 21(1): 35, 2024 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-38622710

RÉSUMÉ

Early breach of the blood-brain barrier (BBB) and consequently extravasation of blood-borne substances into the brain parenchyma is a common hallmark of ischemic stroke. Although BBB breakdown is associated with an increased risk of cerebral hemorrhage and poor clinical prognosis, the cause and mechanism of this process are largely unknown. The aim of this study was to establish an imaging and analysis protocol which enables investigation of the dynamics of BBB breach in relation to hemodynamic properties along the arteriovenous axis. Using longitudinal intravital two-photon imaging following photothrombotic induction of ischemic stroke through a cranial window, we were able to study the response of the cerebral vasculature to ischemia, from the early critical hours to the days/weeks after the infarct. We demonstrate that disruption of the BBB and hemodynamic parameters, including perturbed blood flow, can be studied at single-vessel resolution in the three-dimensional space as early as 30 min after vessel occlusion. Further, we show that this protocol permits longitudinal studies on the response of individual blood vessels to ischemia over time, thus enabling detection of (maladaptive) vascular remodeling such as intussusception, angiogenic sprouting and entanglement of vessel networks. Taken together, this in vivo two-photon imaging and analysis protocol will be useful in future studies investigating the molecular and cellular mechanisms, and the spatial contribution, of BBB breach to disease progression which might ultimately aid the development of new and more precise treatment strategies for ischemic stroke.


Sujet(s)
Encéphalopathie ischémique , Accident vasculaire cérébral ischémique , Accident vasculaire cérébral , Humains , Barrière hémato-encéphalique/métabolisme , Accident vasculaire cérébral/métabolisme , Encéphalopathie ischémique/imagerie diagnostique , Encéphalopathie ischémique/métabolisme , Ischémie/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...