Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 88
Filtrer
1.
Bull Exp Biol Med ; 177(1): 26-29, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38954303

RÉSUMÉ

We present a two-stage model for the study of chronic hind limb ischemia in rats. In the area of ischemia, sclerotic changes with atrophic rhabdomyocytes and reduced vascularization were revealed. CD31 expression in the endothelium increased proportionally to the number of vessels in the ischemic zone, and at the same time, focal expression of ßIII-tubulin was detected in the newly formed nerve fibers. These histological features are equivalent to the development of peripheral arterial disease in humans, which allows using our model in the search for new therapeutic strategies.


Sujet(s)
Modèles animaux de maladie humaine , Membre pelvien , Ischémie , Muscles squelettiques , Animaux , Rats , Muscles squelettiques/anatomopathologie , Muscles squelettiques/métabolisme , Muscles squelettiques/vascularisation , Membre pelvien/vascularisation , Membre pelvien/anatomopathologie , Ischémie/anatomopathologie , Ischémie/métabolisme , Ischémie/physiopathologie , Mâle , Rat Wistar , Antigènes CD31/métabolisme , Tubuline/métabolisme , Maladie artérielle périphérique/anatomopathologie , Maladie artérielle périphérique/métabolisme , Maladie artérielle périphérique/physiopathologie
2.
Dis Model Mech ; 17(5)2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38616715

RÉSUMÉ

Preclinical and clinical studies on the administration of bone marrow-derived cells to restore perfusion show conflicting results. We conducted a systematic review and meta-analysis on preclinical studies to assess the efficacy of bone marrow-derived cells in the hind limb ischemia model and identify possible determinants of therapeutic efficacy. In vivo animal studies were identified using a systematic search in PubMed and EMBASE on 10 January 2022. 85 studies were included for systematic review and meta-analysis. Study characteristics and outcome data on relative perfusion were extracted. The pooled mean difference was estimated using a random effects model. Risk of bias was assessed for all included studies. We found a significant increase in perfusion in the affected limb after administration of bone marrow-derived cells compared to that in the control groups. However, there was a high heterogeneity between studies, which could not be explained. There was a high degree of incomplete reporting across studies. We therefore conclude that the current quality of preclinical research is insufficient (low certainty level as per GRADE assessment) to identify specific factors that might improve human clinical trials.


Sujet(s)
Cellules de la moelle osseuse , Membre pelvien , Ischémie , Animaux , Membre pelvien/vascularisation , Ischémie/thérapie , Ischémie/anatomopathologie , Cellules de la moelle osseuse/cytologie , Perfusion , Transplantation de moelle osseuse , Humains , Biais de publication , Thérapie cellulaire et tissulaire/méthodes
3.
Transl Res ; 270: 13-23, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38548174

RÉSUMÉ

BACKGROUND: Post-ischemic angiogenesis is critical for perfusion recovery and tissue repair. ELABELA (ELA) plays an essential role in embryonic heart development and vasculogenesis. However, the mechanism of ELA on post-ischemic angiogenesis is poorly characterized. METHODS: We first assessed ELA expression after hind limb ischemia (HLI) in mice. We then established a HLI model in tamoxifen-inducible endothelial-ELA-specific knockout mice (ELAECKO) and assessed the rate of perfusion recovery, capillary density, and VEGFR2 pathway. Knockdown of ELA with lentivirus or siRNA and exogenous addition of ELA peptides were employed to analyze the effects of ELA on angiogenic capacity and VEGFR2 pathway in endothelial cells in vitro. The serum levels of ELA in healthy people and patients with type 2 diabetes mellitus (T2DM) and diabetic foot ulcer (DFU) were detected by a commercial ELISA kit. RESULTS: In murine HLI models, ELA was significantly up-regulated in the ischemic hindlimb. Endothelial-specific deletion of ELA impaired perfusion recovery and angiogenesis. In physiologic conditions, no significant difference in VEGFR2 expression was found between ELAECKO mice and ELAWT mice. After ischemia, the expression of VEGFR2, p-VEGFR2, and p-AKT was significantly lower in ELAECKO mice than in ELAWT mice. In cellular experiments, the knockdown of ELA inhibited endothelial cell proliferation and tube formation, and the addition of ELA peptides promoted proliferation and tube formation. Mechanistically, ELA upregulated the expression of VEGFR2, p-VEGFR2, and p-AKT in endothelial cells under hypoxic conditions. In clinical investigations, DFU patients had significantly lower serum levels of ELA compared to T2DM patients. CONCLUSION: Our results indicated that endothelial ELA is a positive regulator of post-ischemic angiogenesis via upregulating VEGFR2 expression. Targeting ELA may be a potential therapeutic option for peripheral arterial diseases.


Sujet(s)
Membre pelvien , Ischémie , Souris knockout , Néovascularisation physiologique , Régulation positive , Récepteur-2 au facteur croissance endothéliale vasculaire , Animaux , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme , Ischémie/métabolisme , Ischémie/génétique , Humains , Souris , Membre pelvien/vascularisation , Mâle , Diabète de type 2/métabolisme , Souris de lignée C57BL , Pied diabétique/métabolisme , Pied diabétique/génétique , Cellules endothéliales/métabolisme , Cellules endothéliales de la veine ombilicale humaine/métabolisme ,
4.
Clin Ther ; 45(11): 1077-1086, 2023 11.
Article de Anglais | MEDLINE | ID: mdl-37770310

RÉSUMÉ

PURPOSE: This narrative review summarizes recent research examining treatment targets for peripheral artery disease (PAD)-related limb ischemia. METHODS: Targeted searches of the PubMed and clinical trial registry databases were performed to identify recent findings from animal models of limb ischemia and clinical studies examining PAD progression and treatment. Ongoing clinical trials testing new treatments for PAD were also reviewed. Relevant full-text articles were retrieved and critically reviewed. Where indicated, data were tabulated and summarized in the text. FINDINGS: Most people with PAD need treatment to improve their walking and function and limit leg pain. Currently, the available treatments of cilostazol, exercise therapy, and revascularization have several deficiencies, including limited access, poor uptake, limited efficacy, and risk of complications. Severe PAD threatens limb viability and is treated by endovascular or open surgical revascularization but is not always successful in achieving limb salvage. Research is ongoing to develop and test new therapies, including new exercise programs, drugs, stem cell treatments and RNA therapeutics, so that new and adjunctive PAD treatments can be offered. Results from multiple clinical trials are expected within the next 5 years. IMPLICATIONS: It is envisaged that a range of new therapies for PAD will be available in the future.


Sujet(s)
Maladie artérielle périphérique , Animaux , Humains , Maladie artérielle périphérique/thérapie , Ischémie/étiologie , Ischémie/thérapie , Sauvetage de membre/effets indésirables , Marche à pied , Résultat thérapeutique , Facteurs de risque
5.
Acta Pharmacol Sin ; 44(12): 2492-2503, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37468692

RÉSUMÉ

Endothelial dysfunction, a central hallmark of cardiovascular pathogenesis in diabetes mellitus, is characterized by impaired endothelial nitric oxide synthase (eNOS) and NO bioavailability. However, the underlying mechanisms remain unclear. Here in this study, we aimed to identify the role of calmodulin (CaM) in diabetic eNOS dysfunction. Human umbilical vein endothelial cells and murine endothelial progenitor cells (EPCs) treated with high glucose (HG) exhibited downregulated CaM mRNA/protein and vascular endothelial growth factor (VEGF) expression with impeded eNOS phosphorylation and cell migration/tube formation. These perturbations were reduplicated in CALM1-knockdown cells but prevented in CALM1-overexpressing cells. EPCs from type 2 diabetes animals behaved similarly to HG-treated normal EPCs, which could be rescued by CALM1-gene transduction. Consistently, diabetic animals displayed impaired eNOS phosphorylation, endothelium-dependent dilation, and CaM expression in the aorta, as well as deficient physical interaction of CaM and eNOS in the gastrocnemius. Local CALM1 gene delivery into a diabetic mouse ischemic hindlimb improved the blunted limb blood perfusion and gastrocnemius angiogenesis, and foot injuries. Diabetic patients showed insufficient foot microvascular autoregulation, eNOS phosphorylation, and NO production with downregulated CaM expression in the arterial endothelium, and abnormal CALM1 transcription in genome-wide sequencing analysis. Therefore, our findings demonstrated that downregulated CaM expression is responsible for endothelium dysfunction and angiogenesis impairment in diabetes, and provided a novel mechanism and target to protect against diabetic endothelial injury.


Sujet(s)
Diabète de type 2 , Humains , Souris , Animaux , Diabète de type 2/métabolisme , Calmoduline/génétique , Calmoduline/métabolisme , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Endothélium/métabolisme , Ischémie/métabolisme , Nitric oxide synthase type III/génétique , Nitric oxide synthase type III/métabolisme , Néovascularisation physiologique
6.
Biol Res ; 56(1): 16, 2023 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-37005678

RÉSUMÉ

BACKGROUND/AIMS: Diabetes mellitus (DM) is highly susceptible to diabetic hind limb ischemia (DHI). MicroRNA (MiR)-17-5p is downregulated in DM and plays a key role in vascular protection. Endothelial progenitor cell (EPC)-released exosomes (EPC-EXs) contribute to vascular protection and ischemic tissue repair by transferring their contained miRs to target cells. Here, we investigated whether miR-17-5p-enriched EPC-EXs (EPC-EXsmiR-17-5p) had conspicuous effects on protecting vascular and skeletal muscle in DHI in vitro and in vivo. METHODS: EPCs transfected with scrambled control or miR-17-5p mimics were used to generate EPC-EXs and EPC-EXsmiR-17-5p. Db/db mice were subjected to hind limb ischemia. After the surgery, EPC-EXs and EPC-EXsmiR-17-5p were injected into the gastrocnemius muscle of the hind limb once every 7 days for 3 weeks. Blood flow, microvessel density, capillary angiogenesis, gastrocnemius muscle weight, structure integrity, and apoptosis in the hind limb were assessed. Vascular endothelial cells (ECs) and myoblast cells (C2C12 cells) were subjected to hypoxia plus high glucose (HG) and cocultured with EPC-EXs and EPC-EXsmiR-17-5p. A bioinformatics assay was used to analyze the potential target gene of miR-17-5p, the levels of SPRED1, PI3K, phosphorylated Akt, cleaved caspase-9 and cleaved caspase-3 were measured, and a PI3K inhibitor (LY294002) was used for pathway analysis. RESULTS: In the DHI mouse model, miR-17-5p was markedly decreased in hind limb vessels and muscle tissues, and infusion of EPC-EXsmiR-17-5p was more effective than EPC-EXs in increasing miR-17-5p levels, blood flow, microvessel density, and capillary angiogenesis, as well as in promoting muscle weight, force production and structural integrity while reducing apoptosis in gastrocnemius muscle. In Hypoxia plus HG-injured ECs and C2C12 cells, we found that EPC-EXsmiR-17-5p could deliver their carried miR-17-5p into target ECs and C2C12 cells and subsequently downregulate the target protein SPRED1 while increasing the levels of PI3K and phosphorylated Akt. EPC-EXsmiR-17-5p were more effective than EPC-EXs in decreasing apoptosis and necrosis while increasing viability, migration, and tube formation in Hypoxia plus HG-injured ECs and in decreasing apoptosis while increasing viability and myotube formation in C2C12 cells. These effects of EPC-EXsmiR-17-5p could be abolished by a PI3K inhibitor (LY294002). CONCLUSION: Our results suggest that miR-17-5p promotes the beneficial effects of EPC-EXs on DHI by protecting vascular ECs and muscle cell functions.


Sujet(s)
Diabète , microARN , Souris , Animaux , Cellules endothéliales , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases , Mouvement cellulaire , Muscles squelettiques/métabolisme , Ischémie , microARN/génétique , microARN/métabolisme , Hypoxie
7.
Front Cardiovasc Med ; 10: 1118738, 2023.
Article de Anglais | MEDLINE | ID: mdl-36937923

RÉSUMÉ

Skeletal muscle injury in peripheral artery disease (PAD) has been attributed to vascular insufficiency, however evidence has demonstrated that muscle cell responses play a role in determining outcomes in limb ischemia. Here, we demonstrate that genetic ablation of Pax7+ muscle progenitor cells (MPCs) in a model of hindlimb ischemia (HLI) inhibited muscle regeneration following ischemic injury, despite a lack of morphological or physiological changes in resting muscle. Compared to control mice (Pax7WT), the ischemic limb of Pax7-deficient mice (Pax7Δ) was unable to generate significant force 7 or 28 days after HLI. A significant increase in adipose was observed in the ischemic limb 28 days after HLI in Pax7Δ mice, which replaced functional muscle. Adipogenesis in Pax7Δ mice corresponded with a significant increase in PDGFRα+ fibro/adipogenic progenitors (FAPs). Inhibition of FAPs with batimastat decreased muscle adipose but increased fibrosis. In vitro, Pax7Δ MPCs failed to form myotubes but displayed increased adipogenesis. Skeletal muscle from patients with critical limb threatening ischemia displayed increased adipose in more ischemic regions of muscle, which corresponded with fewer satellite cells. Collectively, these data demonstrate that Pax7+ MPCs are required for muscle regeneration after ischemia and suggest that muscle regeneration may be an important therapeutic target in PAD.

8.
Biol. Res ; 56: 16-16, 2023. ilus, graf
Article de Anglais | LILACS | ID: biblio-1439483

RÉSUMÉ

BACKGROUND/AIMS: Diabetes mellitus (DM) is highly susceptible to diabetic hind limb ischemia (DHI). MicroRNA (MiR)-17-5p is downregulated in DM and plays a key role in vascular protection. Endothelial progenitor cell (EPC)-released exosomes (EPC-EXs) contribute to vascular protection and ischemic tissue repair by transferring their contained miRs to target cells. Here, we investigated whether miR-17-5p-enriched EPC-EXs (EPC-EXsmiR-17-5p) had conspicuous effects on protecting vascular and skeletal muscle in DHI in vitro and in vivo. METHODS: EPCs transfected with scrambled control or miR-17-5p mimics were used to generate EPC-EXs and EPC-EXsmiR-17-5p. Db/db mice were subjected to hind limb ischemia. After the surgery, EPC-EXs and EPC-EXsmiR-17-5p were injected into the gastrocnemius muscle of the hind limb once every 7 days for 3 weeks. Blood flow, microvessel density, capillary angiogenesis, gastrocnemius muscle weight, structure integrity, and apoptosis in the hind limb were assessed. Vascular endothelial cells (ECs) and myoblast cells (C2C12 cells) were subjected to hypoxia plus high glucose (HG) and cocultured with EPC-EXs and EPC-EXsmiR-17-5p. A bioinformatics assay was used to analyze the potential target gene of miR-17-5p, the levels of SPRED1, PI3K, phosphorylated Akt, cleaved caspase-9 and cleaved caspase-3 were measured, and a PI3K inhibitor (LY294002) was used for pathway analysis. RESULTS: In the DHI mouse model, miR-17-5p was markedly decreased in hind limb vessels and muscle tissues, and infusion of EPC-EXsmiR-17-5p was more effective than EPC-EXs in increasing miR-17-5p levels, blood flow, microvessel density, and capillary angiogenesis, as well as in promoting muscle weight, force production and structural integrity while reducing apoptosis in gastrocnemius muscle. In Hypoxia plus HG-injured ECs and C2C12 cells, we found that EPC-EXsmiR-17-5p could deliver their carried miR-17-5p into target ECs and C2C12 cells and subsequently downregulate the target protein SPRED1 while increasing the levels of PI3K and phosphorylated Akt. EPC-EXsmiR-17-5p were more effective than EPC-EXs in decreasing apoptosis and necrosis while increasing viability, migration, and tube formation in Hypoxia plus HG-injured ECs and in decreasing apoptosis while increasing viability and myotube formation in C2C12 cells. These effects of EPC-EXsmiR-17-5p could be abolished by a PI3K inhibitor (LY294002). CONCLUSION: Our results suggest that miR-17-5p promotes the beneficial effects of EPC-EXs on DHI by protecting vascular ECs and muscle cell functions.


Sujet(s)
Animaux , Souris , microARN/génétique , microARN/métabolisme , Diabète , Mouvement cellulaire , Muscles squelettiques/métabolisme , Phosphatidylinositol 3-kinases , Cellules endothéliales , Ischémie , Hypoxie
9.
Redox Biol ; 56: 102449, 2022 10.
Article de Anglais | MEDLINE | ID: mdl-36063728

RÉSUMÉ

Endothelial progenitor cells (EPCs) are reduced in number and impaired in function in diabetic patients. Whether and how Nrf2 regulates the function of diabetic EPCs remains unclear. In this study, we found that the expression of Nrf2 and its downstream genes were decreased in EPCs from both diabetic patients and db/db mice. Survival ability and angiogenic function of EPCs from diabetic patients and db/db mice also were impaired. Gain- and loss-of-function studies, respectively, showed that knockdown of Nrf2 increased apoptosis and impaired tube formation in EPCs from healthy donors and wild-type mice, while Nrf2 overexpression decreased apoptosis and rescued tube formation in EPCs from diabetic patients and db/db mice. Additionally, proangiogenic function of Nrf2-manipulated mouse EPCs was validated in db/db mice with hind limb ischemia. Mechanistic studies demonstrated that diabetes induced mitochondrial fragmentation and dysfunction of EPCs by dysregulating the abundance of proteins controlling mitochondrial dynamics; upregulating Nrf2 expression attenuated diabetes-induced mitochondrial fragmentation and dysfunction and rectified the abundance of proteins controlling mitochondrial dynamics. Further RNA-sequencing analysis demonstrated that Nrf2 specifically upregulated the transcription of isocitrate dehydrogenase 2 (IDH2), a key enzyme regulating tricarboxylic acid cycle and mitochondrial function. Overexpression of IDH2 rectified Nrf2 knockdown- or diabetes-induced mitochondrial fragmentation and EPC dysfunction. In a therapeutic approach, supplementation of an Nrf2 activator sulforaphane enhanced angiogenesis and blood perfusion recovery in db/db mice with hind limb ischemia. Collectively, these findings indicate that Nrf2 is a potential therapeutic target for improving diabetic EPC function. Thus, elevating Nrf2 expression enhances EPC resistance to diabetes-induced oxidative damage and improves therapeutic efficacy of EPCs in treating diabetic limb ischemia likely via transcriptional upregulating IDH2 expression and improving mitochondrial function of diabetic EPCs.


Sujet(s)
Diabète , Progéniteurs endothéliaux , Animaux , Humains , Souris , Diabète/métabolisme , Progéniteurs endothéliaux/métabolisme , Membre pelvien/métabolisme , Ischémie/métabolisme , Isocitrate dehydrogenases/génétique , Isocitrate dehydrogenases/métabolisme , Dynamique mitochondriale/génétique , Néovascularisation physiologique/génétique , Facteur-2 apparenté à NF-E2/génétique , Facteur-2 apparenté à NF-E2/métabolisme , ARN , Régulation positive
10.
Nutr Metab (Lond) ; 19(1): 59, 2022 Aug 29.
Article de Anglais | MEDLINE | ID: mdl-36038886

RÉSUMÉ

BACKGROUND: The ketogenic diet (KD) has anti-tumor and anti-diabetic effects in addition to its anti-epileptic role. It could also improve cardiac function and attenuate neurological insult. However, the effect of KD on blood perfusion or tissue recovery after ischemia remains largely unknown. Thus, we observed blood flow and ischemic tissue recovery following hind limb ischemia (HLI) in mice. METHODS: C57 mice were fed with either a KD or normal diet (ND) for 2 weeks, before inducing hind limb ischemia, blood perfusion of ischemic limb tissue was observed at 0, 7, and 21 days post operation. RESULTS: KD not only decreased blood perfusion of ischemic limb tissue but also delayed muscle recovery after ischemia, induced muscle atrophy of non-ischemic tissue compared to mice fed with ND. Furthermore, KD delayed wound healing at the surgical site and aggravated inflammation of the ischemic tissue. At the cellular level, KD altered the metabolic status of limb tissue by decreasing glucose and ketone body utilization while increasing fatty acid oxidation. Following ischemia, glycolysis, ketolysis, and fatty acid utilization in limb tissue were all further reduced by KD, while ketogenesis was mildly increased post KD in this mice model. CONCLUSION: The KD may cause impaired tissue recovery after ischemia and possible muscle atrophy under a prolonged diet. Our results hint that patients with limb ischemia should avoid ketogenic diet.

11.
J Am Heart Assoc ; 11(16): e025727, 2022 08 16.
Article de Anglais | MEDLINE | ID: mdl-35946473

RÉSUMÉ

Background Peripheral artery disease is caused by atherosclerotic occlusion of vessels outside the heart and most commonly affects vessels of the lower extremities. Angiogenesis is a part of the postischemic adaptation involved in restoring blood flow in peripheral artery disease. Previously, in a murine hind limb ischemia model of peripheral artery disease, we identified ADAM12 (a disintegrin and metalloproteinase gene 12) as a key genetic modifier of postischemic perfusion recovery. However, less is known about ADAM12 regulation in ischemia. MicroRNAs are a class of small, noncoding, single-stranded RNAs that regulate gene expression primarily through transcriptional repression of messenger RNA (mRNA). We showed microRNA-29a (miR-29a) modulates ADAM12 expression in the setting of diabetes and ischemia. However, how miR-29a modulates ADAM12 is not known. Moreover, the physiological effects of miR-29a modulation in a nondiabetic setting is not known. Methods and Results We overexpressed or inhibited miR-29a in ischemic mouse gastrocnemius and tibialis anterior muscles, and quantified the effect on perfusion recovery, ADAM12 expression, angiogenesis, and skeletal muscle regeneration. In addition, using RNA immunoprecipitation-based anti-miR competitive assay, we investigated the interaction of miR-29a and ADAM12 mRNA in mouse microvascular endothelial cell, skeletal muscle, and human endothelial cell lysates. Ectopic expression of miR-29a in ischemic mouse hind limbs decreased ADAM12 mRNA expression, increased skeletal muscle injury, decreased skeletal muscle function, and decreased angiogenesis and perfusion recovery, with no effect on skeletal muscle regeneration and myofiber cross-sectional area following hind limb ischemia. RNA immunoprecipitation-based anti-miR competitive assay studies showed miR-29a antagomir displaced miR-29a and ADAM12 mRNA from the AGO-2 (Argonaut-2) complex in a dose dependent manner. Conclusions Taken together, the data show miR-29a suppresses ADAM12 expression by directly binding to its mRNA, resulting in impaired skeletal muscle function, angiogenesis, and poor perfusion. Hence, elevated levels of miR-29a, as seen in diabetes and aging, likely contribute to vascular pathology, and modulation of miR-29a could be a therapeutic target.


Sujet(s)
Protéine ADAM12 , microARN , Maladies musculaires , Maladie artérielle périphérique , Protéine ADAM12/génétique , Protéine ADAM12/métabolisme , Animaux , Antagomirs , Humains , Ischémie/métabolisme , Souris , microARN/génétique , microARN/métabolisme , Muscles squelettiques/vascularisation , Néovascularisation physiologique/physiologie , Perfusion , Maladie artérielle périphérique/anatomopathologie , ARN messager/génétique , ARN messager/métabolisme
12.
J Ethnopharmacol ; 292: 115206, 2022 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-35301099

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: Oridonin (Ori), extracted from Isodon rubescens (Hemsl.) H.Hara, is a well-known traditional Chinese herbal medicinal product that possesses antioxidant and anti-inflammatory activities. Oxidative stress and inflammation are the main pathophysiological mechanisms in hindlimb IR injury. However, whether Ori has a protective effect on hind limb IR injury is unknown. AIM OF THE STUDY: The present study was designed to determine the effect of Ori on hindlimb IR injury and its relationship with oxidative stress and inflammation. MATERIALS AND METHODS: The hind limb IR injury model in mice was used to evaluate the protective effect and related mechanisms of Ori. Forty-eight C57BL/6 mice (n = 12 per group) were randomly divided into four groups: Sham group; IR group; IR + Ori (10 mg/kg) group and IR + Ori (20 mg/kg) group. Mice in the IR and IR + Ori groups were subjected to hindlimb IR injury, while mice in the Sham group were subjected to no hindlimb IR injury. HE staining, Masson's staining, TTC staining, DHE staining, TUNEL staining, western blotting analysis and quantitative real-time PCR were employed to explore the mechanisms by which Ori exerts a protective effect on a classical hindlimb IR model in mice. RESULTS: We found that Ori pretreatment prevented muscle damage and decreased cell apoptosis levels compared with the vehicle control. Moreover, the SOD2, CAT, MDA and ROS levels in muscle showed that Ori could significantly reduce oxidative stress in hindlimb IR mice, while the IL-1ß and TNF-α levels in muscle showed that Ori could significantly attenuate IR-induced inflammation. We also found that Ori could increase the expression of Nrf2 and its downstream protein HO-1 and inhibit the expression levels of NLRP3-related proteins (NLRP3, ASC and Caspase-1) in vivo. CONCLUSIONS: Our study suggested that Ori has a protective effect on hindlimb IR injury, which may be related to Nrf2-mediated oxidative stress and NLRP3-mediated inflammasome activation.


Sujet(s)
Facteur-2 apparenté à NF-E2 , Lésion d'ischémie-reperfusion , Animaux , Diterpènes de type kaurane , Membre pelvien , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Souris , Souris de lignée C57BL , Facteur-2 apparenté à NF-E2/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Stress oxydatif , Lésion d'ischémie-reperfusion/métabolisme
13.
Microvasc Res ; 141: 104311, 2022 05.
Article de Anglais | MEDLINE | ID: mdl-34999110

RÉSUMÉ

OBJECTIVES: In the United States, over 8.5 million people suffer from peripheral arterial disease (PAD). Previously we reported that Pellino-1(Peli1) gene therapy reduces ischemic damage in the myocardium and skin flaps in Flk-1 [Fetal Liver kinase receptor-1 (Flk-1)/ Vascular endothelial growth factor receptor-2/VEGFR2] heterozygous (Flk-1+/--) mice. The present study compares the angiogenic response and perfusion efficiency following hind limb ischemia (HLI) in, Flk-1+/- and, MAPKAPKINASE2 (MK2-/-) knockout (KO) mice to their control wild type (WT). We also demonstrated the use of Peli1 gene therapy to improve loss of function following HLI. STUDY DESIGN AND METHODS: Femoral artery ligation (HLI) was performed in both Flk-1+/- and MK2-/- mice along with their corresponding WT. Another set of Flk-1+/- and MK2-/- were injected with either Adeno-LacZ (Ad.LacZ) or Adeno-Peli1 (Ad.Peli1) after HLI. Hind limb perfusion was assessed by laser doppler imaging at specific time points. A standardized scoring scale is used to quantify the extent of ischemia. Histology analysis performed includes capillary density, fibrosis, pro-angiogenic and anti-apoptotic proteins. RESULTS: Flk-1+/- and MK2-/- had a slower recovery of perfusion efficiency in the ischemic limbs than controls. Both Flk-1+/- and MK2-/- KO mice showed decreased capillary density and capillary myocyte ratios with increased fibrosis than their corresponding wild types. Ad.Peli1 injected ischemic Flk-1+/- limb showed improved perfusion, increased capillary density, and pro-angiogenic molecules with reduced fibrosis compared to Ad.LacZ group. No significant improvement in perfusion was observed in MK2-/- ischemic limb after Ad. Peli1 injection. CONCLUSION: Deletion of Flk-1 and MK2 impairs neovascularization and perfusion following HLI. Treatment with Ad. Peli1 results in increased angiogenesis and improved perfusion in Flk-1+/- mice but fails to rectify perfusion in MK2 KO mice. Overall, Peli1 gene therapy is a promising candidate for the treatment of PAD.


Sujet(s)
Maladie artérielle périphérique , Récepteur-2 au facteur croissance endothéliale vasculaire , Animaux , Modèles animaux de maladie humaine , Fibrose , Thérapie génétique/méthodes , Membre pelvien/vascularisation , Humains , Protéines et peptides de signalisation intracellulaire , Ischémie/génétique , Ischémie/anatomopathologie , Ischémie/thérapie , Souris , Souris de lignée C57BL , Souris knockout , Néovascularisation physiologique , Protéines nucléaires/génétique , Perfusion , Maladie artérielle périphérique/génétique , Maladie artérielle périphérique/thérapie , Protein-Serine-Threonine Kinases , Ubiquitin-protein ligases , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme
14.
Angiogenesis ; 25(2): 159-179, 2022 05.
Article de Anglais | MEDLINE | ID: mdl-34524600

RÉSUMÉ

Chemerin is a multifunctional protein initially characterized in our laboratory as a chemoattractant factor for leukocyte populations. Its main functional receptor is CMKLR1. We identified previously chemerin as an anti-tumoral factor inhibiting the vascularization of tumor grafts. We show here that overexpression of bioactive chemerin in mice results in a reduction of the density of the retinal vascular network during its development and in adults. Chemerin did not affect vascular sprouting during the post-natal development of the network, but rather promoted endothelial cell apoptosis and vessel pruning. This phenotype was reversed to normal in CMKLR1-deficient mice, demonstrating the role of this receptor. Chemerin inhibited also neoangiogenesis in a model of pathological proliferative retinopathy, and in response to hind-limb ischemia. Mechanistically, PTEN and FOXO1 antagonists could almost completely restore the density of the retinal vasculature, suggesting the involvement of the PI3-kinase/AKT pathway in the chemerin-induced vessel regression process.


Sujet(s)
Chimiokines , Protéines et peptides de signalisation intercellulaire , Animaux , Apoptose , Chimiokines/métabolisme , Hypoxie , Protéines et peptides de signalisation intercellulaire/génétique , Souris
15.
Molecules ; 26(24)2021 Dec 12.
Article de Anglais | MEDLINE | ID: mdl-34946610

RÉSUMÉ

Critical limb ischemia (CLI) is a severe form of peripheral artery diseases (PAD) and seriously endangers the health of people. Therapeutic angiogenesis represents an important treatment strategy for CLI; various methods have been applied to enhance collateral circulation. However, the current development drug therapy to promote angiogenesis is limited. Resveratrol (RSV), a polyphenol compound extracted from plants, has various properties such as anti-oxidative, anti-inflammatory and anti-cancer effects. Whether RSV exerts protective effects on CLI remains elusive. In the current study, we demonstrated that oral intake of RSV significantly improved hind limb ischemia in mice, and increased the expression of phosphorylated Forkhead box class-O1 (FoxO1). RSV treatment in human umbilical vein endothelial cells (HUVECs) could increase the phosphorylation of FoxO1 and its cytoplasmic re-localization to promote angiogenesis. Then we manipulated FoxO1 in HUVECs to further verify that the effect of RSV on angiogenesis is in a FoxO1-dependent manner. Furthermore, we performed metabolomics to screen the metabolic pathways altered upon RSV intervention. We found that the pathways of pyrimidine metabolism, purine metabolism, as well as alanine, aspartate and glutamate metabolism, were highly correlated with the beneficial effects of RSV on the ischemic muscle. This study provides a novel direction for the medical therapy to CLI.


Sujet(s)
Ischémie chronique menaçant les membres/traitement médicamenteux , Protéine O1 à motif en tête de fourche/métabolisme , Néovascularisation pathologique/traitement médicamenteux , Resvératrol/pharmacologie , Animaux , Ischémie chronique menaçant les membres/métabolisme , Protéine O1 à motif en tête de fourche/génétique , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Humains , Métabolomique , Souris , Souris de lignée C57BL , Néovascularisation pathologique/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques
16.
Antioxidants (Basel) ; 10(11)2021 Oct 27.
Article de Anglais | MEDLINE | ID: mdl-34829572

RÉSUMÉ

Plicosepalus acacia (Fam. Loranthaceae) has been reported to possess hypoglycemic, antioxidant, antimicrobial, and anti-inflammatory effects. Liquid chromatography combined with tandem mass spectrometry (LC-MS/MS) analysis revealed the presence of a high content of polyphenolic compounds that are attributed to the therapeutic effects of the crude extract. In addition, methyl gallate and quercetin were detected as major phytomedicinal agents at concentrations of 1.7% and 0.062 g%, respectively, using high-performance thin layer chromatography (HPTLC). The present study investigated the effect of the P. acacia extract and its isolated compounds, methyl gallate and quercetin, on hind limb ischemia induced in type 1 diabetic rats. Histopathological examination revealed that treatment with P. acacia extract, methyl gallate, and quercetin decreased degenerative changes and inflammation in the ischemic muscle. Further biochemical assessment of the hind limb tissue showed decreased oxidative stress, increased levels of nitric oxide and endothelial nitric oxide synthase (eNOS), and enhancement of the levels of heme oxygenase-1 (HO-1) and vascular endothelial growth factor (VEGF) in the groups treated with methyl gallate and quercetin. Expression levels of hypoxia inducible factor-1 alpha (HIF-1α), VEGF, fibroblast growth factor-2 (FGF-2), and miR-146a were upregulated in the muscle tissue of methyl gallate- and quercetin-treated groups along with downregulation of nuclear factor kappa B (NF-κB). In conclusion, P. acacia extract and its isolated compounds, methyl gallate and quercetin, mediated therapeutic angiogenesis in diabetic hind limb ischemia.

17.
J Mol Histol ; 52(6): 1155-1164, 2021 Dec.
Article de Anglais | MEDLINE | ID: mdl-34642827

RÉSUMÉ

Cell-based therapeutics bring great hope in areas of unmet medical needs. Mesenchymal stem cells (MSCs) have been suggested to facilitate neovascularization mainly by paracrine action. Endothelial progenitor cells (EPCs) can migrate to ischemic sites and participate in angiogenesis. The combination cell therapy that includes MSCs and EPCs has a favorable effect on ischemic limbs. However, the mechanism of combination cell therapy remains unclear. Herein, we investigate whether stromal cell-derived factor (SDF)-1 secreted by MSCs contributes to EPC migration to ischemic sites via CXCR4/Phosphoinositide 3-Kinases (PI3K)/protein kinase B (termed as AKT) signaling pathway. First, by a "dual-administration" approach, intramuscular MSC injections were supplemented with intravenous Qdot® 525 labeled-EPC injections in the mouse model of hind limb ischemia. Then, the mechanism of MSC effect on EPC migration was detected by the transwell system, tube-like structure formation assays, western blot assays in vitro. Results showed that the combination delivery of MSCs and EPCs enhanced the incorporation of EPCs into the vasculature and increased the capillary density in mouse ischemic hind limb. The numbers of CXCR4-positive EPCs increased after incubation with MSC-conditioned medium (CM). MSCs contributed to EPC migration and tube-like structure formation, both of which were suppressed by AMD3100 and wortmannin. Phospho-AKT induced by MSC-CM was attenuated when EPCs were pretreated with AMD3100 and wortmannin. In conclusion, we confirmed that MSCs contributes to EPC migration, which is mediated via CXCR4/PI3K/AKT signaling pathway.


Sujet(s)
Chimiokine CXCL12/biosynthèse , Progéniteurs endothéliaux/métabolisme , Cellules souches mésenchymateuses/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Récepteurs CXCR4/métabolisme , Transduction du signal , Animaux , Communication cellulaire , Mouvement cellulaire , Cellules cultivées , Modèles animaux de maladie humaine , Membres/vascularisation , Membres/anatomopathologie , Immunophénotypage , Ischémie/étiologie , Ischémie/métabolisme , Ischémie/anatomopathologie , Ischémie/thérapie , Transplantation de cellules souches mésenchymateuses/méthodes , Souris , Néovascularisation physiologique/génétique , Phosphorylation
18.
J Control Release ; 338: 773-783, 2021 10 10.
Article de Anglais | MEDLINE | ID: mdl-34530052

RÉSUMÉ

Pro-angiogenic growth factors have been studied as potential therapeutics for cardiovascular diseases like critical limb ischemia (CLI). However, the translation of these factors has remained a challenge, in part, due to problems associated with safe and effective delivery. Here, we describe a hydrogel-based delivery system for growth factors where release is modulated by focused ultrasound (FUS), specifically a mechanism termed acoustic droplet vaporization. With these fibrin-based, acoustically-responsive scaffolds (ARSs), release of a growth factor is non-invasively and spatiotemporally-controlled in an on-demand manner using non-thermal FUS. In vitro studies demonstrated sustained release of basic fibroblast growth factor (bFGF) from the ARSs using repeated applications of FUS. In in vivo studies, ARSs containing bFGF were implanted in mice following induction of hind limb ischemia, a preclinical model of CLI. During the 4-week study, mice in the ARS + FUS group longitudinally exhibited significantly more perfusion and less visible necrosis compared to other experimental groups. Additionally, significantly greater angiogenesis and less fibrosis were observed for the ARS + FUS group. Overall, these results highlight a promising, FUS-based method of delivering a pro-angiogenic growth factor for stimulating angiogenesis and reperfusion in a cardiovascular disease model. More broadly, these results could be used to personalize the delivery of therapeutics in different regenerative applications by actively controlling the release of a growth factor.


Sujet(s)
Fibrine , Facteur de croissance fibroblastique de type 2 , Animaux , Membre pelvien , Hydrogels , Ischémie/thérapie , Souris , Néovascularisation physiologique , Volatilisation
19.
Nagoya J Med Sci ; 83(3): 465-476, 2021 Aug.
Article de Anglais | MEDLINE | ID: mdl-34552283

RÉSUMÉ

Adipose-derived regenerative cells (ADRCs), mesenchymal stem/progenitor cells from subcutaneous adipose tissue, have been shown to stimulate angiogenesis in hind limb ischemia, an effect attributed to paracrine action on endothelial cells (ECs) in mice. Despite promising therapeutic effects, the relevant molecules promoting neovascularization in this setting have not been fully elucidated. Extracellular vesicles, crucial mediators of intercellular communication, are recognized as a new therapeutic modality for regenerative medicine. Here, we found that GW4869, an exosome biogenesis inhibitor targeting neutral sphingomyelinase, impaired ADRCs-mediated angiogenesis and improvement of blood perfusion in a murine hind limb ischemia model. In addition, while the supernatant of ADRCs induced murine EC migration, this effect was attenuated by pre-treatment with GW4869. RNA analysis revealed that treatment of ADRCs with GW4869 reduced the expression of microRNA-21 (miR-21), miR-27b, miR-322, and let-7i in ADRCs-derived exosomes. Furthermore, the exosomes derived from GW4869-treated ADRCs induced the expression of the miR-21 targets Smad7 and Pten, and the miR-322 target Cul2, in ECs. These findings suggest that several miRNAs in ADRCs-derived exosomes contribute to angiogenesis and improvement of blood perfusion in a murine hind limb ischemia model.


Sujet(s)
Ischémie , Tissu adipeux , Animaux , Cellules endothéliales , Ischémie/thérapie , Souris , microARN/génétique , Néovascularisation pathologique
20.
Vascul Pharmacol ; 141: 106906, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34509635

RÉSUMÉ

BACKGROUND: Pro-angiogenic microRNA modulation is a potentially attractive approach in the management of peripheral artery disease (PAD). The aim of this systematic review and meta-analysis was to examine the impact of microRNAs involved in the process of angiogenesis on blood flow recovery following hind limb ischemia induction in animal models. METHODS: A literature search was performed to identify studies testing the efficacy of microRNA treatment on animal models of hind limb ischemia. Following that, a meta-analysis of the included studies was executed with the primary outcome being the change in ischemic-to-normal hind limb perfusion ratio assessed via laser Doppler imaging. Moreover, risk of bias, sensitivity analysis and publication bias were evaluated. RESULTS: Studies evaluation led to the inclusion of 18 studies whose meta-analysis suggested that microRNA treatment resulted in improved ischemic hind limb perfusion 7 [standardized mean difference (SMD): 0.93, 95% CI 0.49-1.38], 14 (SMD: 1.31, 95% CI 0.78-1.84), and 21 days (SMD: 1.13, 95% CI 0.59-1.66) after hind limb ischemia induction. Moderate-to-substantial heterogeneity and possible publication bias were noted. Risk of bias was unclear despite the balanced baseline animal characteristics. CONCLUSION: The present meta-analysis suggests that pro-angiogenic modulation of microRNAs accelerates vascular perfusion recovery in animal models of acute hind limb ischemia. Further studies on animal models with similar characteristics to that of PAD patients are warranted to translate those findings in human PAD setting.


Sujet(s)
microARN , Maladie artérielle périphérique , Animaux , Modèles animaux de maladie humaine , Membre pelvien/vascularisation , Humains , Ischémie , microARN/génétique , Muscles squelettiques/vascularisation , Néovascularisation physiologique , Maladie artérielle périphérique/génétique , Débit sanguin régional
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE