Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Microvasc Res ; 139: 104253, 2022 01.
Article in English | MEDLINE | ID: mdl-34520773

ABSTRACT

Chalcones and sulfonamides are well-known chemical groups associated with several biological activities such as antibiotic, anti-inflammatory, and antitumor activities. Over the past few decades, a series of sulfonamide-chalcone hybrids have been synthesized and assessed to develop compounds with interesting biological properties for application in disease therapy. In the present study, a new sulfonamide-chalcone hybrid µ - (2,5-dichloro-N-{4-[(3E)-4-(3-nitrophenyl) buta-1,3-dien-2-yl] phenyl} benzene sulfonamide), or simply CL185, was synthesized, and its angiogenic activity was assessed using the chick embryo chorioallantoic membrane (CAM) assay at different concentrations (12.5, 25, and 50 µg/µL). To further investigate the role of CL185 in the angiogenic process, we evaluated the levels of vascular endothelial growth factor (VEGF) in all treated CAMs. The results showed that all concentrations of CL185 significantly increased tissue vascularization (p < 0.05) as well as the parameters associated with angiogenesis, in which inflammation was the most marked phenomenon observed. In all CAMs treated with CL185, VEGF levels were significantly higher than those in the negative control (p < 0.05), and at the highest concentration, VEGF levels were even higher than in the positive control (p < 0.05). The pronounced angiogenic activity displayed by CL185 may be related to the increase in VEGF levels that were stimulated by inflammatory processes observed in our study. Therefore, CL185 presents a favorable profile for the development of drugs that can be used in pro-angiogenic and tissue repair therapies.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Chalcones/pharmacology , Chorioallantoic Membrane/blood supply , Inflammation/metabolism , Neovascularization, Physiologic/drug effects , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inducing Agents/toxicity , Animals , Chalcones/toxicity , Chick Embryo , Inflammation/chemically induced , Up-Regulation
2.
J Ethnopharmacol ; 273: 113824, 2021 Jun 12.
Article in English | MEDLINE | ID: mdl-33581257

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Buxuhuayu decoction (BXHYD) has been frequently used to treat patients with diabetic ulcers (DUs), without notable adverse reactions. However, the related molecular mechanism remains unelucidated. AIM OF THE STUDY: This study assessed the potential mechanism of BXHYD against DUs by using network pharmacology and animal experiments. MATERIALS AND METHODS: First, high-performance liquid chromatography (HPLC) was used for quality control of BXHYD. Further, the hub compounds and targets were screened from the Active Compound-Targets (ACT) network and the protein and protein interaction (PPI) network. Enrichment analysis was performed using DAVID, and molecular docking technology was used to identify active compounds that may play a key role in pub targets. Finally, a DUs animal model was established and used to elucidate the effect of BXHYD on the PI3K/Akt/eNOS signalling pathway. RESULTS: (1) Calycosin-7-glucoside, amygdalin, and tanshinone iiA were detected in the freeze-dried powder of BXHYD. (2) Twelve hub compounds and eight hub targets were screened using the ACT and PPI networks. Through molecular docking, it was found that the four hub targets (TP53, IL6, VEGFA, and AKT1) binds luteolin and quercetin more tightly. (3) BXHYD is most likely to promote angiogenesis and wound healing by activating the PI3K/Akt/eNOS signalling pathway. CONCLUSIONS: This research revealed that BXHYD might activate the PI3K/Akt/eNOS signalling pathway to promote DUs healing. These findings support the clinical use of BXHYD and provide the foundation for its subsequent studies.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Neovascularization, Physiologic/drug effects , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ulcer/drug therapy , Angiogenesis Inducing Agents/chemistry , Angiogenesis Inducing Agents/therapeutic use , Angiogenesis Inducing Agents/toxicity , Animals , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/therapeutic use , Drugs, Chinese Herbal/toxicity , Male , Medicine, Chinese Traditional , Molecular Docking Simulation , Protein Interaction Maps , Rats, Sprague-Dawley , Signal Transduction/drug effects , Streptozocin , Ulcer/etiology , Ulcer/pathology , Wound Healing/drug effects
3.
Biochem Pharmacol ; 189: 114192, 2021 07.
Article in English | MEDLINE | ID: mdl-32783891

ABSTRACT

Exposure to toxic metal contaminants, such as cadmium compounds (Cd2+), has been shown to induce adverse effects on various organs and tissues. In particular, blood vessels are severely impacted by Cd2+ exposure, which may lead to cardiovascular diseases (CVDs). According to previous studies, CVDs are associated with increased cyclooxygenase 2 (COX-2) levels. However, the mechanisms by which CdCl2-induced COX-2 overexpression leads to cardiovascular dysfunction remain unclear. Herein, we show that the relative gene expressions of VEGF and PTGS2 (COX-2 encoding gene) are positively correlated in CVDs patients. Moreover, we demonstrate that the in vitro administration of CdCl2 induces cytotoxicity and endoplasmic reticulum (ER) stress in primary human umbilical vein endothelial cells (HUVECs). The induction of ER stress and the overexpression of COX-2 in CdCl2-treated cells alters the protein level of vascular endothelial growth factor (VEGF), resulting in abnormal angiogenesis and increased cytotoxicity. At the pre-transcription level, the inhibition of ER stress by siGRP78 (a key mediator of ER stress) can restore normal angiogenesis in the CdCl2-exposed cells. Meanwhile, at the transcription level, the adverse effects of CdCl2 exposure may be reversed via genetic modification with siRNA (siPTGS2) or by using phytochemical inhibitors (parthenolide, PN) of COX-2. Finally, at the post-transcription level, COX-2 expression may be restricted by the binding of microRNA-101 (miR-101) to the 3'-UTR of PTGS2 mRNA. The use of mimic miR-101 (mi101) to induce the expression of miR-101 eventually leads to reduced COX-2 protein levels, relieved ER stress, and less abnormal angiogenesis and cytotoxicity of CdCl2-exposed primary HUVECs. Overall, our results suggest that CdCl2-induced abnormal angiogenesis is mediated by miR-101/COX-2/VEGF-axis-dependent ER stress, and that cardiovascular dysfunction may be controlled by manipulating COX-2 at the pre-transcription, transcription, and post-transcription levels.


Subject(s)
Angiogenesis Inducing Agents/toxicity , Cadmium Chloride/toxicity , Cyclooxygenase 2/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , MicroRNAs/metabolism , Cell Survival/drug effects , Cell Survival/physiology , Cyclooxygenase 2 Inhibitors/administration & dosage , Dose-Response Relationship, Drug , Human Umbilical Vein Endothelial Cells/metabolism , Humans
4.
Int J Pharm ; 576: 118997, 2020 Feb 25.
Article in English | MEDLINE | ID: mdl-31893542

ABSTRACT

Cardiovascular diseases (CVD) are the leading cause of death worldwide. Growth factor therapy has emerged as novel therapeutic strategy under investigation for CVD. In this sense, adrenomedullin-2 (ADM-2) has been recently identified as a new angiogenic factor able to regulate the regional blood flow and cardiovascular function. However, the therapeutic value of ADM-2 is limited by its short biological half-life and low plasma stability. Poly (lactic-co-glycolic acid) (PLGA) micro- and nanoparticles have been investigated as growth factor delivery systems for cardiac repair. In this study, we aimed to develop PLGA nanoparticles containing ADM-2 intended for therapeutic angiogenesis. PLGA nanoparticles containing ADM-2 were prepared by a double emulsion modified method, resulting in 300 nm-sized stable particles with zeta potential around - 30 mV. Electron microscopy analysis by SEM and TEM revealed spherical particles with a smooth surface. High encapsulation efficiency was reached (ca.70%), as quantified by ELISA. ADM-2 associated to polymer nanoparticles was also determined by EDS elemental composition analysis, SDS-PAGE and LC-MS/MS for peptide identification. In vitro release assays showed the sustained release of ADM-2 from polymer nanoparticles for 21 days. Cell viability experiments were performed in J774 macrophages and H9c2 cardiomyocyte cells, about which PLGA nanoparticles loaded with ADM-2 did not cause toxicity in the range 0.01-1 mg/ml. Of note, encapsulated ADM-2 significantly induced cell proliferation in EA.hy926 endothelial cells, indicating the ADM-2 bioactivity was preserved after the encapsulation process. Collectively, these results demonstrate the feasibility of using PLGA nanoparticles as delivery systems for the angiogenic peptide ADM-2, which could represent a novel approach for therapeutic angiogenesis in CVD using growth factor therapy.


Subject(s)
Angiogenesis Inducing Agents/administration & dosage , Cell Proliferation/drug effects , Drug Carriers , Endothelial Cells/drug effects , Peptide Hormones/administration & dosage , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Angiogenesis Inducing Agents/chemistry , Angiogenesis Inducing Agents/toxicity , Animals , Cell Line , Delayed-Action Preparations , Drug Compounding , Drug Liberation , Humans , Kinetics , Mice , Nanoparticles , Peptide Hormones/chemistry , Peptide Hormones/toxicity , Polylactic Acid-Polyglycolic Acid Copolymer/toxicity , Rats , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Solubility
5.
J Cardiovasc Pharmacol ; 74(1): 71-79, 2019 07.
Article in English | MEDLINE | ID: mdl-31274845

ABSTRACT

Ganglioside has been implicated to play important roles in modulating various cell signaling and biological functions. However, the functional analysis of a single ganglioside in a zebrafish model is so far lacking. In this study, we investigated the angiogenic effects of 2 monosialoganglioside compounds isolated from GM1 in zebrafish embryos. First, we showed the tested compounds are adequate safe. Then, we found that these compounds exhibited significant proangiogenic effect through enhancement of endothelial cell proliferation, migration, and differentiation. Furthermore, the 2 compounds were proved to promote angiogenesis through, at least partially, modulating the level of Notch signaling. This study provides the novel insights into the clinical application of the 2 ganglioside compounds and GM1.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Embryo, Nonmammalian/blood supply , Endothelial Cells/drug effects , G(M1) Ganglioside/pharmacology , Neovascularization, Physiologic/drug effects , Angiogenesis Inducing Agents/toxicity , Animals , Animals, Genetically Modified , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Endothelial Cells/metabolism , G(M1) Ganglioside/toxicity , Gene Expression Regulation, Developmental , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
6.
Biomed Pharmacother ; 103: 844-850, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29710500

ABSTRACT

Vascular smooth muscle cell (VSMC) proliferation and migration play critical roles in the development and progression of atherosclerosis. C1q/tumor necrosis factor-related protein 6 (CTRP6), a member of CTRPs family, was involved in cardiovascular diseases, inflammatory reaction and adipogenesis. However, the role of CTRP6 in VSMCs remains largely unknown. The purpose of this study is to investigate the effects of CTRP6 on VSMC proliferation and migration and explore the possible mechanism. Our results indicated that CTRP6 expression was dramatically down-regulated in human atherosclerotic tissues and in cultured VSMCs stimulated by platelet-derived growth factor-BB (PDGF-BB). In addition, CTRP6 overexpression significantly inhibited the proliferation and migration of VSMCs exposed to PDGF-BB, as well as increased expression of α-SMA and SM22α in PDGF-BB-stimulated VSMCs. Furthermore, CTRP6 overexpression efficiently prevented the activation of PI3K/Akt/mTOR in VSMCs in response to PDGF-BB. In conclusion, these findings showed that CTRP6 inhibits PDGF-BB-induced VSMC proliferation and migration, at least in part, through suppressing the PI3K/Akt/mTOR signaling pathway. Therefore, CTRP6 may be a potential target for the treatment of atherosclerosis.


Subject(s)
Angiogenesis Inducing Agents/toxicity , Cell Movement/physiology , Cell Proliferation/physiology , Collagen/biosynthesis , Muscle, Smooth, Vascular/metabolism , Proto-Oncogene Proteins c-sis/toxicity , Becaplermin , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Humans , Muscle, Smooth, Vascular/drug effects
7.
Kidney Int ; 93(4): 842-854, 2018 04.
Article in English | MEDLINE | ID: mdl-29273331

ABSTRACT

We recently developed a therapeutic biopolymer composed of an elastin-like polypeptide (ELP) fused to vascular endothelial growth factor (VEGF) and showed long-term renoprotective effects in experimental renovascular disease after a single intra-renal administration. Here, we sought to determine the specificity, safety, efficacy, and mechanisms of renoprotection of ELP-VEGF after systemic therapy in renovascular disease. We tested whether kidney selectivity of the ELP carrier would reduce off-target binding of VEGF in other organs. In vivo bio-distribution after systemic administration of ELP-VEGF in swine was determined in kidneys, liver, spleen, and heart. Stenotic-kidney renal blood flow and glomerular filtration rate were quantified in vivo using multi-detector computed tomography (CT) after six weeks of renovascular disease, then treated with a single intravenous dose of ELP-VEGF or placebo and observed for four weeks. CT studies were then repeated and the pigs euthanized. Ex vivo studies quantified renal microvascular density (micro-CT) and fibrosis. Kidneys, liver, spleen, and heart were excised to quantify the expression of angiogenic mediators and markers of progenitor cells. ELP-VEGF accumulated predominantly in the kidney and stimulated renal blood flow, glomerular filtration rate, improved cortical microvascular density, and renal fibrosis, and was accompanied by enhanced renal expression of VEGF, downstream mediators of VEGF signaling, and markers of progenitor cells compared to placebo. Expression of angiogenic factors in liver, spleen, and heart were not different compared to placebo-control. Thus, ELP efficiently directs VEGF to the kidney after systemic administration and induces long-term renoprotection without off-target effects, supporting the feasibility and safety of renal therapeutic angiogenesis via systemic administration of a novel kidney-specific bioengineered compound.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Kidney/blood supply , Kidney/drug effects , Neovascularization, Physiologic/drug effects , Peptides/pharmacology , Renal Artery Obstruction/drug therapy , Vascular Endothelial Growth Factor A/pharmacology , Angiogenesis Inducing Agents/administration & dosage , Angiogenesis Inducing Agents/pharmacokinetics , Angiogenesis Inducing Agents/toxicity , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Disease Models, Animal , Drug Carriers , Fibrosis , Glomerular Filtration Rate/drug effects , Injections, Intravenous , Kidney/metabolism , Kidney/pathology , Peptides/administration & dosage , Peptides/pharmacokinetics , Peptides/toxicity , Recombinant Fusion Proteins/pharmacology , Renal Artery Obstruction/metabolism , Renal Artery Obstruction/pathology , Renal Artery Obstruction/physiopathology , Renal Circulation/drug effects , Stem Cells/drug effects , Stem Cells/metabolism , Sus scrofa , Tissue Distribution , Vascular Endothelial Growth Factor A/administration & dosage , Vascular Endothelial Growth Factor A/pharmacokinetics , Vascular Endothelial Growth Factor A/toxicity
8.
J Cardiovasc Transl Res ; 10(4): 348-358, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28397162

ABSTRACT

Angiogenic growth factor therapy for ischemic cardiovascular disease carries a risk of stimulating atherosclerotic plaque growth. We evaluated risk benefit ratio of sustained administration of recombinant human placental growth factor (rhPlGF)-2 in mice with advanced atherosclerosis and chronic ischemic cardiomyopathy. We maintained apolipoprotein E-deficient mice on a high cholesterol diet and induced myocardial infarction by transient ligation at 4 weeks. At 8 weeks, we assessed left ventricular (LV) function and randomized mice to receive rhPlGF-2 or vehicle (VEH) subcutaneously for 28 days. Administration of rhPlGF-2 significantly increased PlGF plasma levels without adverse hemodynamic or systemic inflammatory effects. RhPlGF-2 did not increase plaque area, composition, or vulnerability in the aortic arch. RhPlGF-2 significantly improved contractile function and reduced LV end-systolic and end-diastolic volume indices with a concomitant increase in capillary and arteriolar density in ischemic myocardium. RhPlGF-2 may represent a promising therapeutic strategy in chronic ischemic cardiomyopathy.


Subject(s)
Angiogenesis Inducing Agents/administration & dosage , Aorta/drug effects , Aortic Diseases/drug therapy , Atherosclerosis/drug therapy , Cardiomyopathies/drug therapy , Myocardial Infarction/drug therapy , Neovascularization, Physiologic/drug effects , Placenta Growth Factor/administration & dosage , Ventricular Function, Left/drug effects , Angiogenesis Inducing Agents/toxicity , Animals , Aorta/pathology , Aorta/physiopathology , Aortic Diseases/pathology , Aortic Diseases/physiopathology , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/physiopathology , Cholesterol, Dietary , Chronic Disease , Disease Models, Animal , Infusions, Subcutaneous , Male , Mice, Knockout, ApoE , Myocardial Contraction/drug effects , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/physiopathology , Placenta Growth Factor/toxicity , Plaque, Atherosclerotic , Recombinant Proteins/administration & dosage , Recovery of Function , Stroke Volume/drug effects , Time Factors , Vascular Stiffness/drug effects , Ventricular Remodeling/drug effects
9.
Microvasc Res ; 108: 29-33, 2016 11.
Article in English | MEDLINE | ID: mdl-27418582

ABSTRACT

The search for new bioactive molecules is a driving force for research pharmaceutical industries, especially those molecules obtained from fermentation. The molecules possessing angiogenic and anti-inflammatory attributes have attracted attention and are the focus of this study. Angiogenic activity from kefir polysaccharide extract, via chorioallantoic membrane assay, exhibited a pro-angiogenic effect compared with vascular endothelial factor (pro-angiogenic) and hydrocortisone (anti-angiogenic) activity as standards with an EC50 of 192ng/mL. In terms of anti-inflammatory activity determined via hyaluronidase enzyme assay, kefir polysaccharide extract inhibited the enzyme with a minimal activity of 2.08mg/mL and a maximum activity of 2.57mg/mL. For pharmaceutical purposes, kefir polysaccharide extract is considered to be safe because it does not inhibit VERO cells in cytotoxicity assays.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Chorioallantoic Membrane/blood supply , Glycoside Hydrolase Inhibitors/pharmacology , Hyaluronoglucosaminidase/antagonists & inhibitors , Kefir/microbiology , Neovascularization, Physiologic/drug effects , Polysaccharides, Bacterial/pharmacology , Angiogenesis Inducing Agents/isolation & purification , Angiogenesis Inducing Agents/toxicity , Animals , Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/toxicity , Cell Survival/drug effects , Chick Embryo , Chickens , Chlorocebus aethiops , Dose-Response Relationship, Drug , Glycoside Hydrolase Inhibitors/isolation & purification , Glycoside Hydrolase Inhibitors/toxicity , Hyaluronoglucosaminidase/metabolism , Polysaccharides, Bacterial/isolation & purification , Polysaccharides, Bacterial/toxicity , Vero Cells
10.
Biol Trace Elem Res ; 166(1): 57-65, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25893364

ABSTRACT

Powder mixtures of tungsten carbide and metallic cobalt (WC-Co) are widely used in various products. Nanoparticles are engineered structures with at least one dimension of 100 nm or smaller. WC-Co is known to be associated with lung injury and diseases. Angiogenesis is a key process during vasculature, carcinogenesis, recovery of injury, and inflammatory diseases. However, the cellular effects of WC-Co nanoparticles on angiogenesis remain to be elucidated. In this study, we investigated angiogenic response and relative mechanisms after exposure to WC-Co nanoparticles. Our results showed that WC-Co nanoparticles at 5 µg/cm(2) induced ROS production which activated AKT and ERK1/2 signaling pathways in lung epithelial cells by reactive oxygen species (ROS) staining and immunoblotting; WC-Co treatment also increased transcriptional activation of AP-1, NF-κB, and VEGF by reporter assay. Further studies demonstrated that ROS are upstream molecules of AKT and ERK signaling pathways; the activation of AP-1, NF-κB, and VEGF was through ROS generation, AKT and ERK1/2 activation. In addition, WC-Co nanoparticles affected the cells to induce angiogenesis by chicken chorioallantoic membrane (CAM) assay. These results illustrate that exposure to WC-Co nanoparticles induces angiogenic response by activating ROS, AKT, and ERK1/2 signaling pathways and the downstream molecules and elucidate the potential molecular mechanisms during this process. This information may be useful for preventing potential damage from nanoparticle exposure in the future.


Subject(s)
Angiogenesis Inducing Agents/toxicity , Cobalt/toxicity , MAP Kinase Signaling System/drug effects , NF-kappa B/metabolism , Nanoparticles/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Transcription Factor AP-1/metabolism , Tungsten Compounds/toxicity , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inducing Agents/chemistry , Animals , Cell Line, Tumor , Chickens , Chorioallantoic Membrane/blood supply , Cobalt/chemistry , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Luciferases/genetics , MAP Kinase Signaling System/genetics , NF-kappa B/genetics , Proto-Oncogene Proteins c-akt/genetics , Transcription Factor AP-1/genetics , Transcriptional Activation/drug effects , Transfection , Tungsten Compounds/chemistry , Vascular Endothelial Growth Factor A/genetics
11.
Vascul Pharmacol ; 60(3): 110-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24534483

ABSTRACT

Wogonin, a natural monoflavonoid extracted from Scutellariae radix, has been reported for its ability of inhibiting tumor angiogenesis. In this study, we assessed the effect of wogonin on angiogenesis induced by low level of H2O2 (10 µM) in human umbilical vein endothelial cells (HUVECs). Wogonin suppressed H2O2-induced migration and tube formation of HUVECs as well as microvessel sprouting from rat aortic rings in vitro. Meanwhile, wogonin suppressed vessel growth in chicken chorioallantoic membrane (CAM) model in vivo. Mechanistic studies showed that wogonin suppressed H2O2-activated PI3K/Akt pathway and reduced the expression of vascular endothelial growth factor (VEGF) up-regulated by H2O2 in both protein and mRNA levels. In addition, wogonin also inhibited nuclear translocation of NF-κB, and decreased the binding ability of NF-κB with exogenous consensus DNA oligonucleotide. Then we further investigated the effect of wogonin on over-activated PI3K/Akt pathway by insulin-like growth factor-1 (IGF-1) and H2O2. We found that wogonin suppressed phosphorylation of Akt, up-regulation of VEGF and angiogenesis in vitro which was further induced by IGF-1 and H2O2. Moreover, in NF-κB overexpressed HUVECs, wogonin could also reduce the expression of VEGF and inhibited the migration and tube formation. Taken together, these results suggested that wogonin was potential in inhibiting H2O2-induced angiogenesis in vitro and in vivo via suppressing PI3K/Akt pathway and NF-κB signaling.


Subject(s)
Angiogenesis Inducing Agents , Flavanones/pharmacology , Hydrogen Peroxide/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Angiogenesis Inducing Agents/toxicity , Animals , Chick Embryo , Dose-Response Relationship, Drug , Human Umbilical Vein Endothelial Cells , Humans , Hydrogen Peroxide/toxicity , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Scutellaria baicalensis , Signal Transduction/drug effects , Signal Transduction/physiology
12.
Am J Physiol Renal Physiol ; 305(7): F1031-41, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23926180

ABSTRACT

In the present study, we examine the hypothesis that the nitric oxide (NO) produced by endothelial NO synthase (eNOS) plays a protective role in the development of ANG II-induced hypertension and renal injury by minimizing oxidative stress and the inflammation induced by TNF-α. Systolic blood pressure (SBP) and renal injury responses to chronic infusions of ANG II (via implanted minipumps) were evaluated for 2 wk in wild-type (WT) and in eNOS knockout mice (KO) cotreated with or without a superoxide (O2(-)) scavenger, tempol (400 mg/l in the drinking water), or a TNF-α receptor blocker, etanercept (5 mg/kg/day ip). In study 1, when ANG II was given at a dose of 25 ng/min, it increased mean SBP in WT mice (Δ36 ± 3 mmHg; n = 7), and this effect was attenuated in mice pretreated with tempol (Δ24 ± 3 mmHg; n = 6). In KO mice (n = 9), this dose of ANG II resulted in severe renal injury associated with high mortality. To avoid this high mortality in KO, study 2 was conducted with a lower dose of ANG II (10 ng/min) that increased SBP slightly in WT (Δ17 ± 7 mmHg; n = 6) but exaggeratedly in KO (Δ48 ± 12 mmHg, n = 6) associated with severe renal injury. Cotreatment with either tempol (n = 6) or etanercept (n = 6) ameliorated the hypertensive, as well as the renal injury responses in KO compared with WT. These data demonstrate a protective role for eNOS activity in preventing renal inflammatory injury and hypertension induced by chronic increases in ANG II.


Subject(s)
Angiotensin II/physiology , Hypertension/enzymology , Hypertension/prevention & control , Nephritis/enzymology , Nitric Oxide Synthase Type III/physiology , Ribonuclease, Pancreatic/toxicity , Angiogenesis Inducing Agents/toxicity , Angiotensin II/administration & dosage , Animals , Hypertension/etiology , Inflammation/enzymology , Inflammation/pathology , Male , Mice , Mice, Knockout , Nephritis/etiology , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Up-Regulation/drug effects , Up-Regulation/physiology
13.
J Ethnopharmacol ; 139(2): 447-53, 2012 Jan 31.
Article in English | MEDLINE | ID: mdl-22138660

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Diabetic foot ulceration results in high risk of lower extremity amputation, and represents a significant health care expenditure worldwide. Radix Astragali (RA) and Radix Rehmanniae (RR) are widely used Chinese medicinal herbs in treating diabetes, and have shown positive effects in enhancing wound healing in diabetic foot ulcer animal model. MATERIALS AND METHODS: The angiogenic efficacy of NF3, a simplified 2-herb formula consisting of RA and RR in 2:1 ratio, was investigated. Median lethal concentration (LC50) and median effective concentration (EC50) were determined by treating zebrafish embryos with different concentrations of NF3 from 20 hpf to 72 hpf. The angiogenic activity of NF3 was examined in zebrafish embryos in vivo and by rat aortic ring assay in vitro. Cell cycle analysis of endothelial cells induced by NF3 was analyzed by flow cytometry using transgenic zebrafish Tg(fli1:EGFP). Real-time PCR was used to analyze mRNA expression profiles of selected genes involved in VEGF, FGF and MAPK pathways. RESULTS: NF3 enhanced blood vessel formation as indicated by extra growth of intersegmental vessels in zebrafish embryos, and increased microvessels formation in rat aortic ring. NF3 also enhanced endothelial cells proliferation as shown by increased percentage of cells accumulating in S phase and G2/M phase of the cell cycle. NF3 exposure significantly induced up-regulation of VEGF-A, Flk-1, fgf1 and bRaf expression in zebrafish embryos. CONCLUSIONS: Our results demonstrated that NF3 was effective in promoting angiogenesis in zebrafish embryos and by rat aortic ring assay, which provided scientific basis to support the use of NF3 as potential therapeutics in treating diabetic foot ulceration.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Aorta/drug effects , Drugs, Chinese Herbal/pharmacology , Endothelial Cells/drug effects , Neovascularization, Physiologic/drug effects , Angiogenesis Inducing Agents/therapeutic use , Angiogenesis Inducing Agents/toxicity , Angiogenic Proteins/genetics , Animals , Animals, Genetically Modified , Cell Proliferation/drug effects , Diabetic Foot/drug therapy , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/therapeutic use , Drugs, Chinese Herbal/toxicity , Embryo, Nonmammalian/blood supply , Embryo, Nonmammalian/drug effects , Endothelial Cells/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation/drug effects , Humans , Lethal Dose 50 , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Male , Neovascularization, Physiologic/genetics , Proto-Oncogene Protein c-fli-1/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , S Phase Cell Cycle Checkpoints/drug effects , Zebrafish/embryology , Zebrafish/genetics
14.
Toxicology ; 288(1-3): 34-42, 2011 Oct 09.
Article in English | MEDLINE | ID: mdl-21762757

ABSTRACT

Due to the toxic effect of asbestos, other materials with similar chemical-physical characteristics have been introduced to substitute it. We evaluate the angiogenic effect of certain asbestos substitute fibres such as glass fibres (GFs), ceramic fibres (CFs) and wollastonite fibres (WFs) and then compare angiogenic responses to those induced by crocidolite asbestos fibres (AFs). An in vitro model using human endothelial cells in small islands within a culture matrix of fibroblasts (Angio-Kit) was used to evaluate vessel formation. The release of IL-6, sIL-R6, IL-8, VEGF-A and their soluble receptors, sVEGFR-1, sVEGFR-2, was determined in the conditioning medium of Angio-Kit system after fibre treatment. ROS formation and cell viability were evaluated in cultured endothelial cells (HUVEC). To evaluate the involvement of intracellular mechanisms, EGFR signalling, ROS formation and nuclear factor-κB (NFκB) pathway were then inhibited by incubating HUVEC cells with AG1478, NAC and PDTC respectively, and the cytokine and growth factor release was analyzed in the culture medium after 7 days of fibre incubation. Among the mineral fibres tested, WFs markedly induced blood vessel formation which was associated with release of IL-6 and IL-8, VEGF-A and their soluble receptors. ROS production was observed in HUVEC after WFs treatment which was associated with cell cytotoxicity. The EGFR-induced ERK phosphorylation and ROS-mediated NFκB activation were involved in the cytokine and angiogenic factor release. However, only the EGFR activation was able to induce angiogenesis. The WFs are potential angiogenic agents that can induce regenerative cytokine and angiogenic factor production resulting in the formation of new blood vessels.


Subject(s)
Angiogenesis Inducing Agents/toxicity , Endothelial Cells/drug effects , Mineral Fibers/toxicity , Neovascularization, Pathologic/chemically induced , Neovascularization, Physiologic/drug effects , Angiogenesis Inducing Agents/chemistry , Asbestos, Crocidolite/toxicity , Calcium Compounds/toxicity , Cell Survival/drug effects , Cells, Cultured , Ceramics/toxicity , Cytokines/drug effects , Cytokines/metabolism , Endothelial Cells/metabolism , ErbB Receptors/metabolism , Fibroblasts/metabolism , Glass , Humans , Reactive Oxygen Species/metabolism , Silicates/toxicity , Umbilical Cord , Vascular Endothelial Growth Factor A/metabolism
15.
Curr Eye Res ; 36(9): 857-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21599469

ABSTRACT

PURPOSE: To describe the presence of iris neovascularization in a rabbit-model of retinal neovascularization induced by the intravitreal injection of latex-derived angiogenic fraction microspheres (LAF). MATERIALS AND METHODS: Eight New Zealand rabbits received one intravitreal injection of PLGA (L-lactide-co-glycolide) microspheres with 50 ug of LAF in the right eye (Group A). Microspheres without the LAF (0.1 ml) were injected in controls (Group B; n = 8). Follow-up with clinical evaluation and iris fluorescein angiography was performed after 4 weeks when eyes were processed for light microscopy. RESULTS: All eyes from Group A showed significant vascular dilation, conjunctival hyperemia and neovascularization on the iris surface, after LAF injection. No vascular changes were observed in Group B. CONCLUSIONS: The intravitreal injection of microspheres containing the LAF can induce rubeosis iridis in rabbits and could be used as a simple experimental model for iris neovascularization.


Subject(s)
Angiogenesis Inducing Agents/toxicity , Glaucoma, Neovascular/etiology , Iris/blood supply , Latex/toxicity , Neovascularization, Pathologic/chemically induced , Angiogenesis Inducing Agents/administration & dosage , Animals , Disease Models, Animal , Disease Progression , Drug Carriers , Female , Fluorescein Angiography , Fundus Oculi , Glaucoma, Neovascular/pathology , Intravitreal Injections , Iris/drug effects , Lactic Acid , Latex/administration & dosage , Microspheres , Neovascularization, Pathologic/complications , Neovascularization, Pathologic/pathology , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Rabbits , Risk Factors
16.
Int J Toxicol ; 29(5): 496-504, 2010.
Article in English | MEDLINE | ID: mdl-20884859

ABSTRACT

Peroxisome proliferator-activated receptor α (PPARα) agonism in ocular inflammation has not been thoroughly investigated. The objective of this investigation was to determine the effect of WY-14 643, a selective PPARα agonist, on inflammatory cytokine release in human ocular cells. Stimulation of primary human corneal epithelial cells, keratocytes, and retinal endothelial cells with 1 to 10 ng/mL interleukin 1ß (IL-1ß) resulted in a significant increase in numerous inflammatory cytokines, including IL-6, IL-8, and tumor necrosis factor α (TNF-α); and dexamethasone was able to significantly inhibit these effects. However, WY-14 643 did not effectively block IL-1ß-induced cytokine release in ocular cells; rather, significant increases in IL-1ß-induced inflammatory cytokines were observed in these cells but not in aortic smooth muscle cells. WY-14 643 also significantly upregulated vascular endothelial growth factor (VEGF) expression in corneal epithelial cells and keratocytes. These studies demonstrate for the first time that PPARα agonism may be proinflammatory and proangiogenic in a variety of ocular cells and suggest that therapeutic applications of such agents in ophthalmology may be limited.


Subject(s)
Eye/drug effects , Eye/metabolism , Inflammation Mediators/metabolism , PPAR alpha/agonists , Peroxisome Proliferators/toxicity , Pyrimidines/toxicity , Angiogenesis Inducing Agents/toxicity , Cells, Cultured , Colony-Stimulating Factors/metabolism , Cytokines/metabolism , Epithelium, Corneal/cytology , Epithelium, Corneal/drug effects , Epithelium, Corneal/metabolism , Eye/cytology , Humans , Inflammation/chemically induced , Inflammation/metabolism , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Interleukins/metabolism , Organ Specificity , PPAR alpha/genetics , PPAR alpha/metabolism , RNA, Messenger/metabolism , Retina/cytology , Retina/drug effects , Retina/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factors/metabolism
17.
Toxicol Appl Pharmacol ; 244(3): 291-9, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20083128

ABSTRACT

Angiogenesis and vessel remodeling are fundamental to the pathogenesis of a number of diseases caused by environmental arsenic exposure, including tumorigenesis and cardiovascular diseases. Arsenic (AsIII) has been shown to stimulate angiogenesis and vascular remodeling in vivo. However, the exact molecular mechanisms accounting for arsenic-induced angiogenesis are not clear. The present study investigates the role of heme oxygenase-1 (HO-1) in sodium arsenite-mediated angiogenesis in vitro. Transwell assay, three-dimensional Matrigel assay, RT-PCR, ELISA and immunoblotting were used to determine cell migration, vascular tube formation, mRNA and protein expression. Chromatin immunoprecipitation and luciferase assay were applied to examine the DNA binding with protein and HO-1 transcriptional activity. Here, we report that low concentrations of arsenite (0.1-1 muM) stimulated cell migration and vascular tube formation in human microvascular endothelial cells (HMVEC). Arsenite induced HO-1 mRNA and protein expression. Knock down of HO-1 expression decreased arsenite-induced VEGF expression, cell migration, and tube formation. We showed that arsenite promoted dissociation of Bach1 (a transcriptional repressor) from the HO-1 enhancers and increased Nrf2 binding to these elements. Site directed mutagenesis assay identified that Bach1 cysteine residues 557 and 574 were essential for the induction of HO-1 gene in response to arsenite. These findings demonstrate a role for HO-1 in arsenite-mediated angiogenesis in vitro.


Subject(s)
Angiogenesis Inducing Agents/toxicity , Arsenic/toxicity , Heme Oxygenase-1/metabolism , Neovascularization, Physiologic/drug effects , Arsenites/toxicity , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Movement/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fanconi Anemia Complementation Group Proteins/metabolism , Gene Expression Regulation/drug effects , Humans , MAP Kinase Kinase 4/metabolism , Microvessels/drug effects , Microvessels/metabolism , NF-E2-Related Factor 2/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/metabolism , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Vascular Endothelial Growth Factor A/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Curr Eye Res ; 35(1): 56-62, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20021255

ABSTRACT

PURPOSE: To create a retinal neovascularization experimental model using intravitreal injection of microspheres loaded with latex-derived angiogenic fraction. METHODS: Thirty-two albino New Zealand rabbits, divided in 4 groups of 8 animals, were enrolled in this study. Rabbits in groups I, II, and III received one intravitreal injection of PLGA (L-lactide-co-glycolide) microspheres with 10, 30, and 50 microg of latex-derived angiogenic fraction into their right eyes, respectively, and group IV received 0.1 ml of microspheres without the angiogenic fraction. Weekly follow-up with ophthalmoscopy and fluorescein angiography was performed; the rabbits were sacrificed in the 4th week and their eyes processed for light microscopy. RESULTS: All eyes from group I demonstrated increased retinal vascular tortuosity, observed from 14 days after injection and maintained for 28 days, otherwise without new vessels detection. All group II eyes showed vascular changes similar to group I. Fifty percent of the eyes from group II rabbits developed retinal neovascularization 21 days after injection. All eyes from group III demonstrated significant vascular tortuosity and retinal new vessels 2 weeks after injection, progressing to fibrovascular proliferation and tractional retinal detachment. No vascular changes or retinal new vessels were observed in group IV eyes. Light microscopy confirmed the existence of new vessels previously seen on fluorescein angiography, in retinal sections adjacent to the optic disc, not observed in sections at the same area in the control group. CONCLUSION: Thirty- and 50-microg microspheres containing latex-derived angiogenic fraction injected into the vitreous cavity induced retinal neovascularization in rabbits.


Subject(s)
Angiogenesis Inducing Agents/toxicity , Disease Models, Animal , Latex/toxicity , Retinal Neovascularization/chemically induced , Retinal Vessels/drug effects , Animals , Drug Carriers , Female , Fluorescein Angiography , Lactic Acid , Microspheres , Ophthalmoscopy , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Rabbits , Retinal Neovascularization/diagnosis , Retinal Vessels/pathology
19.
J Vasc Res ; 46(2): 103-8, 2009.
Article in English | MEDLINE | ID: mdl-18617751

ABSTRACT

We have previously demonstrated that iloprost, a stable prostacyclin (PGI(2)) analogue, induces angiogenesis in vivo, through a vascular endothelial growth factor (VEGF)-dependent mechanism. In this study, we demonstrate that iloprost-induced angiogenesis and VEGF upregulation are modulated by peroxisome proliferator-activated receptor-alpha (PPARalpha), a ligand-inducible transcription factor that belongs to the nuclear hormone receptor superfamily and plays multiple biological activities in the vascular system. We show that iloprost is unable to induce angiogenesis in mice lacking the PPARalpha gene (PPARalpha-/- mice). Likewise, iloprost-induced VEGF upregulation is absent in PPARalpha-/- mice. In contrast, iloprost induces a robust angiogenic response in wild-type mice, along with local upregulation of VEGF. Importantly, mice lacking the PPARalpha gene exhibit a normal angiogenic response to VEGF, indicating that the absence of PPARalpha does not result in a general impairment of angiogenesis, but specifically affects the ability of iloprost to induce angiogenesis. Our data demonstrate unexpected functional relationships between the PGI(2) system and the PPAR signaling pathway and shed new light on the molecular mechanisms involved in iloprost-induced angiogenesis.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Corneal Neovascularization/chemically induced , Iloprost/pharmacology , PPAR alpha/drug effects , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inducing Agents/toxicity , Animals , Corneal Neovascularization/metabolism , Iloprost/toxicity , Mice , Mice, Knockout , PPAR alpha/deficiency , PPAR alpha/genetics , PPAR alpha/metabolism , RNA, Messenger/metabolism , Signal Transduction/drug effects , Transfection , Up-Regulation , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism
20.
Arterioscler Thromb Vasc Biol ; 27(12): 2548-54, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17932321

ABSTRACT

OBJECTIVE: Hypoxia inducible factor (HIF) plays a pivotal role in the adaptation to ischemic conditions. Its activity is modulated by an oxygen-dependent hydroxylation of proline residues by prolyl hydroxylases (PHD). METHODS AND RESULTS: We discovered 2 unique compounds (TM6008 and TM6089), which inhibited PHD and stabilized HIF activity in vitro. Our docking simulation studies based on the 3-dimensional structure of human PHD2 disclosed that they preferentially bind to the active site of PHD. Whereas PHD inhibitors previously reported inhibit PHD activity via iron chelation, TM6089 does not share an iron chelating motif and is devoid of iron chelating activity. In vitro Matrigel assays and in vivo sponge assays demonstrated enhancement of angiogenesis by local administration of TM6008 and TM6089. Their oral administration stimulated HIF activity in various organs of transgenic rats expressing a hypoxia-responsive reporter vector. No acute toxicity was observed up to 2 weeks after a single oral dose of 2000 mg/kg for TM6008. Oral administration of TM6008 protected neurons in a model of cerebrovascular disease. The protection was associated with amelioration of apoptosis but independent of enhanced angiogenesis. CONCLUSIONS: The present study uncovered beneficial effects of novel PHD inhibitors preferentially binding to the active site of PHD.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Brain Ischemia/prevention & control , Enzyme Inhibitors/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neovascularization, Physiologic/drug effects , Neuroprotective Agents/pharmacology , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Administration, Oral , Angiogenesis Inducing Agents/administration & dosage , Angiogenesis Inducing Agents/chemistry , Angiogenesis Inducing Agents/pharmacokinetics , Angiogenesis Inducing Agents/toxicity , Animals , Animals, Genetically Modified , Apoptosis/drug effects , Binding Sites , Brain Ischemia/pathology , Cells, Cultured , Chelating Agents/pharmacology , Computer Simulation , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Design , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/toxicity , Genes, Reporter , Gerbillinae , Heart/drug effects , Humans , Hydroxylation , Hypoxia-Inducible Factor-Proline Dioxygenases , Kidney/drug effects , Kidney/metabolism , Lethal Dose 50 , Liver/drug effects , Liver/metabolism , Models, Molecular , Molecular Structure , Myocardium/metabolism , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/toxicity , Procollagen-Proline Dioxygenase/chemistry , Procollagen-Proline Dioxygenase/metabolism , Protein Binding , Protein Conformation , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...