Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Med Sci Monit ; 27: e928153, 2021 Mar 26.
Article in English | MEDLINE | ID: mdl-33770068

ABSTRACT

BACKGROUND The primary cause of death in patients with diabetes mellitus (DM) is diabetic macroangiopathy, a complication that related to the function and number of endothelial progenitor cells (EPCs). Salvianic acid A (SAA) is a water-soluble active ingredient of Salvia miltiorrhiza, a traditional Chinese medicine used to treat cardiovascular diseases. The purpose of this study was to explore the effects of SAA on the function of rat EPCs cultured in vitro in a high-glucose environment. MATERIAL AND METHODS Bone marrow-derived EPCs from 40 Sprague-Dawley rats were identified by fluorescence staining. Cell viability, apoptosis, tube formation, lactated dehydrogenase (LDH) release, and nitric oxide (NO) production were detected by 3-[4,5-dimethylthylthiazol-2-yl]-2,5 diphenyltetrazolium bromide assay, flow cytometry, tube formation, LDH, and 3-amino,4-aminomethyl-2',7'-difluorescein, and diacetate assays, respectively. The expression levels of proteins were examined by western blotting. RESULTS Cultured EPCs showed a cobblestone morphology and positive expression of Dil-ac-LDL and FITC-UEA-1. High glucose impaired cell viability. Different concentrations of SAA had no significant effect on EPC viability. SAA reduced the apoptosis rate and LDH release, but promoted tube formation, viability, and NO production in high-glucose-treated EPCs. The ratios of p-AKT/AKT and p-eNOS/eNOS in high-glucose-treated EPCs were elevated by SAA. Phosphoinositide 3-kinase inhibitor LY294002 blocked the rescue effects of SAA on high-glucose-treated EPCs. CONCLUSIONS SAA protected EPCs against high-glucose-induced dysfunction via the AKT/eNOS pathway.


Subject(s)
Diabetes Mellitus/drug therapy , Endothelial Cells/physiology , Endothelial Progenitor Cells/physiology , Lactates/metabolism , Medicine, Chinese Traditional , Nitric Oxide Synthase Type III/metabolism , Animals , Cell Survival , Cells, Cultured , Glucose/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Salvia , Signal Transduction
2.
Phytomedicine ; 61: 152850, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31035054

ABSTRACT

BACKGROUD: Endothelial progenitor cells (EPCs) have been characterized as one of the key effectors of endothelial healing. The effect of Danhong injection (DHI), the most widely prescribed Chinese medicine for coronary heart disease (CHD), on EPCs mobilization remains unclear. PURPOSE: We aimed to assess the effect of DHI on EPCs mobilization to repair percutaneous coronary intervention (PCI) induced vascular injury, and to investigate the characteristics and potential mechanism of DHI on EPCs mobilization. METHOD: Forty-two patients with CHD underwent PCI and received stent implantation were enrolled in a Phase II clinical trials. All patients received routine western medical treatment after PCI, patients of DHI group received DHI in addition. The levels of CECs, cytokines (vWF, IL-6, CRP) and EPCs were analyzed at baseline, post-PCI and after treatment. To investigate the characteristics of DHI on EPCs mobilization, 12 healthy volunteers received intravenous infusion of DHI once and the other 12 received for 7 days. EPCs enumeration were done at a series of time points. At last we tested the effect of DHI and three chemical constituents of DHI (danshensu; lithospermic acid, LA; salvianolic acid D, SaD) on EPCs level and expression of Akt, eNOS and MMP-9 in bone marrow cells of myocardial infarction (MI) mice. RESULTS: In the DHI group the angina symptoms were improved, the levels of cytokines and CECs were reduced; while EPCs population was increased after treatment. In the phase I clinical trials, EPCs counts reached a plateau phase in 9 h and maintained for more than 10 h after a single dose. After continuous administration, EPCs levels plateaued on the 3rd or 4th day, and maintain till 1 day after the withdrawal, then its levels gradually declined. DHI treatment induced a timely dependent mobilization of EPCs. DHI promoted EPCs mobilization via upregulating the expression of Akt, eNOS and MMP-9 in BM. LA and SaD have played a valuable role in EPCs mobilization. CONCLUSION: These initial results demonstrated that DHI is effective in alleviating endothelial injury and promoting endothelial repair through enhancing EPCs mobilization and revealed the effect feature and possible mechanisms of DHI in mobilizing EPCs.


Subject(s)
Cardiovascular Agents/pharmacology , Coronary Disease/drug therapy , Coronary Disease/surgery , Drugs, Chinese Herbal/pharmacology , Endothelial Progenitor Cells/drug effects , Endothelium, Vascular/injuries , Aged , Animals , Cardiovascular Agents/administration & dosage , Drugs, Chinese Herbal/administration & dosage , Endothelial Progenitor Cells/physiology , Female , Humans , Injections , Male , Matrix Metalloproteinase 9/metabolism , Mice, Inbred C57BL , Middle Aged , Myocardial Infarction/drug therapy , Nitric Oxide Synthase Type III/metabolism , Percutaneous Coronary Intervention/adverse effects , Proto-Oncogene Proteins c-akt/metabolism , Vascular System Injuries/drug therapy , Vascular System Injuries/etiology
3.
Sci China Life Sci ; 61(10): 1178-1188, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30159681

ABSTRACT

The rapid endothelialization of tissue-engineered blood vessels (TEBVs) can effectively prevent thrombosis and inhibit intimal hyperplasia. The traditional Chinese medicine ingredient icariin is highly promising for the treatment of cardiovascular diseases. ß-cyclodextrin sulfate is a type of hollow molecule that has good biocompatibility and anticoagulation properties and exhibits a sustained release of icariin. We studied whether icariin-loaded ß-cyclodextrin sulfate can promote the endothelialization of TEBVs. The experimental results showed that icariin could significantly promote the proliferation and migration of endothelial progenitor cells; at the same time, icariin could promote the migration of rat vascular endothelial cells (RAVECs). Subsequently, we used an electrostatic force to modify the surface of the TEBVs with icariin-loaded ß-cyclodextrin sulfate, and these vessels were implanted into the rat common carotid artery. After 3 months, micro-CT results showed that the TEBVs modified using icariin-loaded ß-cyclodextrin sulfate had a greater patency rate. Scanning electron microscopy (SEM) and CD31 immunofluorescence results showed a better degree of endothelialization. Taken together, icariin-loaded ß-cyclodextrin sulfate can achieve anticoagulation and rapid endothelialization of TEBVs to ensure their long-term patency.


Subject(s)
Blood Coagulation/drug effects , Blood Vessels/drug effects , Endothelial Progenitor Cells/drug effects , Flavonoids/pharmacology , beta-Cyclodextrins/pharmacology , Animals , Blood Vessel Prosthesis , Blood Vessels/metabolism , Blood Vessels/physiology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/physiology , Flavonoids/chemistry , Rats, Sprague-Dawley , Sulfates/metabolism , Tissue Engineering/methods , beta-Cyclodextrins/chemistry , beta-Cyclodextrins/metabolism
4.
Am J Clin Nutr ; 107(6): 876-882, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29741564

ABSTRACT

Background: Endothelial progenitor cells (EPCs) and microparticles are emerging as novel markers of cardiovascular disease (CVD) risk, which could potentially be modified by dietary fat. We have previously shown that replacing dietary saturated fatty acids (SFAs) with monounsaturated or n-6 (ω-6) polyunsaturated fatty acids (MUFAs or PUFAs, respectively) improved lipid biomarkers, blood pressure, and markers of endothelial activation, but their effects on circulating EPCs and microparticles are unclear. Objective: The Dietary Intervention and VAScular function (DIVAS) Study investigated the replacement of 9.5-9.6% of total energy (%TE) contributed by SFAs with MUFAs or n-6 PUFAs for 16 wk on EPC and microparticle numbers in United Kingdom adults with moderate CVD risk. Design: In this randomized, controlled, single-blind, parallel-group dietary intervention, men and women aged 21-60 y (n = 190) with moderate CVD risk (≥50% above the population mean) consumed 1 of three 16-wk isoenergetic diets. Target compositions for total fat, SFAs, MUFAs, and n-6 PUFAs (%TE) were as follows: SFA-rich diet (36:17:11:4; n = 64), MUFA-rich diet (36:9:19:4; n = 62), and n-6 PUFA-rich diet (36:9:13:10; n = 66). Circulating EPC, endothelial microparticle (EMP), and platelet microparticle (PMP) numbers were analyzed by flow cytometry. Dietary intake, vascular function, and other cardiometabolic risk factors were determined at baseline. Results: Relative to the SFA-rich diet, MUFA- and n-6 PUFA-rich diets decreased EMP (-47.3%, -44.9%) respectively and PMP (-36.8%, -39.1%) numbers (overall diet effects, P < 0.01). The MUFA-rich diet increased EPC numbers (+28.4%; P = 0.023). Additional analyses that used stepwise regression models identified the augmentation index (measuring arterial stiffness determined by pulse-wave analysis) as an independent predictor of baseline EPC and microparticle numbers. Conclusions: Replacement of 9.5-9.6%TE dietary SFAs with MUFAs increased EPC numbers, and replacement with either MUFAs or n-6 PUFAs decreased microparticle numbers, suggesting beneficial effects on endothelial repair and maintenance. Further studies are warranted to determine the mechanisms underlying the favorable effects on EPC and microparticle numbers after SFA replacement. This trial was registered at www.clinicaltrials.gov as NCT01478958.


Subject(s)
Cardiovascular Diseases/blood , Cardiovascular Diseases/prevention & control , Cell-Derived Microparticles/physiology , Dietary Fats/administration & dosage , Endothelial Progenitor Cells/physiology , Adult , Biomarkers , Cohort Studies , Dietary Fats/classification , Fatty Acids/administration & dosage , Fatty Acids, Unsaturated/administration & dosage , Female , Humans , Male , Middle Aged , Single-Blind Method , Young Adult
5.
Medicine (Baltimore) ; 97(6): e9736, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29419665

ABSTRACT

INTRODUCTION: Septic shock is a potentially fatal organ dysfunction caused by an imbalance of the host response to infection. The changes in microcirculation during sepsis can be explained by the alterations in the endothelial barrier function. Endothelial progenitor cells (EPCs) are a potential recovery index of endothelial function and it an increase in response to neuromuscular electrical stimulation (NMES) was demonstrated. Therefore, the objective of this study is to investigate the effects of NMES in patients with septic shock. METHODS AND ANALYSIS: It is a study protocol for a randomized cross-over design in an intensive care unit of a tertiary University hospital. Thirty-one patients aged 18 to 65 years. The study will be divided in 2 phases: the phase one will be held in the first 72 hours of septic shock and the phase two after 3 days of first assessment. Patients will be randomly selected to the intervention protocol (decubitus position with the limbs raised and NMES) and control protocol (decubitus position with the limbs raised without NMES). After this procedure, the patients will be allocated in group 1 (intervention and control protocol) or group 2 (control and intervention protocol) with a wash-out period of 4 to 6 hours between them. The main outcome is mobilization of EPCs. The secondary outcome is metabolic and hemodynamic data. A linear mixed model will be used for analysis of dependent variables and estimated values of the mean of the differences of each effect.


Subject(s)
Endothelial Progenitor Cells/physiology , Multiple Organ Failure/prevention & control , Shock, Septic/therapy , Transcutaneous Electric Nerve Stimulation/methods , Adult , Critical Care/methods , Cross-Over Studies , Female , Humans , Male , Microcirculation/physiology , Middle Aged , Multiple Organ Failure/etiology , Outcome Assessment, Health Care , Research Design , Shock, Septic/complications
6.
Clin Cancer Res ; 23(3): 666-676, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27769988

ABSTRACT

PURPOSE: Bone marrow-derived progenitor cells, including VEGFR2+ endothelial progenitor cells (EPCs) and copper-dependent pathways, model the tumor microenvironment. We hypothesized that copper depletion using tetrathiomolybdate would reduce EPCs in high risk for patients with breast cancer who have relapsed. We investigated the effect of tetrathiomolybdate on the tumor microenvironment in preclinical models. EXPERIMENTAL DESIGN: Patients with stage II triple-negative breast cancer (TNBC), stage III and stage IV without any evidence of disease (NED), received oral tetrathiomolybdate to maintain ceruloplasmin (Cp) between 8 and 17 mg/dL for 2 years or until relapse. Endpoints were effect on EPCs and other biomarkers, safety, event-free (EFS), and overall survival (OS). For laboratory studies, MDA-LM2-luciferase cells were implanted into CB17-SCID mice and treated with tetrathiomolybdate or water. Tumor progression was quantified by bioluminescence imaging (BLI), copper depletion status by Cp oxidase levels, lysyl oxidase (LOX) activity by ELISA, and collagen deposition. RESULTS: Seventy-five patients enrolled; 51 patients completed 2 years (1,396 cycles). Most common grade 3/4 toxicity was neutropenia (3.7%). Lower Cp levels correlated with reduced EPCs (P = 0.002) and LOXL-2 (P < 0.001). Two-year EFS for patients with stage II-III and stage IV NED was 91% and 67%, respectively. For patients with TNBC, EFS was 90% (adjuvant patients) and 69% (stage IV NED patients) at a median follow-up of 6.3 years, respectively. In preclinical models, tetrathiomolybdate decreased metastases to lungs (P = 0.04), LOX activity (P = 0.03), and collagen crosslinking (P = 0.012). CONCLUSIONS: Tetrathiomolybdate is safe, well tolerated, and affects copper-dependent components of the tumor microenvironment. Biomarker-driven clinical trials in high risk for patients with recurrent breast cancer are warranted. Clin Cancer Res; 23(3); 666-76. ©2016 AACR.


Subject(s)
Adenocarcinoma/secondary , Breast Neoplasms/drug therapy , Chelating Agents/therapeutic use , Copper/metabolism , Endothelial Progenitor Cells/drug effects , Lung Neoplasms/secondary , Molybdenum/therapeutic use , Tumor Microenvironment/drug effects , Adenocarcinoma/drug therapy , Adenocarcinoma/prevention & control , Amino Acid Oxidoreductases/blood , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Ceruloplasmin/analysis , Chelating Agents/pharmacology , Disease Progression , Disease-Free Survival , Endothelial Progenitor Cells/physiology , Female , Follow-Up Studies , Humans , Lung Neoplasms/prevention & control , Mice, SCID , Molybdenum/pharmacology , Neoplasm Proteins/blood , Neovascularization, Pathologic/physiopathology , Neovascularization, Pathologic/prevention & control , Neutropenia/chemically induced , Risk , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
7.
PLoS One ; 10(9): e0138832, 2015.
Article in English | MEDLINE | ID: mdl-26398523

ABSTRACT

Although high density lipoprotein (HDL) improves the functions of endothelial progenitor cells (EPCs), the effect of HDL ApoAI mimetic peptide reverse-D-4F (Rev-D4F) on EPC mobilization and repair of EPC dysfunctions remains to be studied. In this study, we investigated the effects of Rev-D4F on peripheral blood cell subpopulations in C57 mice treated with a high fat diet and the mechanism of Rev-D4F in improving the function of EPCs impaired by tumor necrosis factor-α (TNF-α). The high fat diet significantly decreased the number of EPCs, EPC migratory functions, and the percentage of lymphocytes in the white blood cells. However, it significantly increased the number of white blood cells, the percentage of monocytes in the white blood cells, and the level of vascular endothelial growth factor (VEGF) and TNF-α in the plasma. Rev-D4F clearly inhibited the effect of the high fat diet on the quantification of peripheral blood cell subpopulations and cytokine levels, and increased stromal cell derived factor 1α (SDF-1α) in the plasma. We provided in vitro evidence that TNF-α impaired EPC proliferation, migration, and tube formation through inactive AKT and eNOS, which was restored by Rev-D4F treatment. In contrast, both the PI3-kinase (PI3K) inhibitor (LY294002) and AKT inhibitor (perifosine) obviously inhibited the restoration of Rev-4F on EPCs impaired by TNF-α. Our results suggested that Rev-D4F increases the quantity of endothelial progenitor cells through increasing the SDF-1α levels and decreasing the TNF-α level of peripheral blood in high fat diet-induced C57BL/6J mice, and restores TNF-α induced dysfunctions of EPCs partly through stimulating the PI3K/AKT signal pathway.


Subject(s)
Cardiovascular Diseases/pathology , Diet, High-Fat/adverse effects , Endothelial Progenitor Cells/physiology , Peptides/pharmacology , Animals , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Cells, Cultured , Chemokine CXCL12/metabolism , Drug Evaluation, Preclinical , Endothelial Progenitor Cells/drug effects , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
Molecules ; 20(9): 17557-69, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26402665

ABSTRACT

Andrographis paniculata (A. paniculata, Chuanxinlian), a medicinal herb with an extremely bitter taste that is native to China and other parts of Southeast Asia, possesses immense therapeutic value; however, its therapeutic properties have rarely been applied in the field of skin care. In this study, we investigated the effect of an A. paniculata extract (APE) on human epidermal stem cells (EpSCs), and confirmed its anti-aging effect through in vitro, ex vivo, and in vivo study. An MTT assay was used to determine cell proliferation. A flow cytometric analysis, with propidium iodide, was used to evaluate the cell cycle. The expression of integrin ß1 (CD29), the stem cell marker, was detected with antibodies, using flow cytometry in vitro, and immunohistochemical assays in ex vivo. Type 1 collagen and VEGF (vascular endothelial growth factor) were measured using an enzyme-linked immunosorbent assay (ELISA). During the clinical study, skin hydration, elasticity, wrinkling, sagging, and dermal density were evaluated before treatment and at four and eight weeks after the treatment with the test product (containing the APE) on the face. The proliferation of the EpSCs, treated with the APE, increased significantly. In the cell cycle analysis, the APE increased the G2/M and S stages in a dose-dependent manner. The expression of integrin ß1, which is related to epidermal progenitor cell expansion, was up-regulated in the APE-treated EpSCs and skin explants. In addition, the production of VEGF in the EpSCs increased significantly in response to the APE treatment. Consistent with these results, the VEGF and APE-treated EpSCs conditioned medium enhanced the Type 1 collagen production in normal human fibroblasts (NHFs). In the clinical study, the APE improved skin hydration, dermal density, wrinkling, and sagging significantly. Our findings revealed that the APE promotes a proliferation of EpSCs, through the up-regulation of the integrin ß1 and VEGF expression. The VEGF might affect the collagen synthesis of NHF as a paracrine factor. Clinical studies further suggested that treatment with formulations containing APE confers anti-aging benefits. Based on these results, we suggest that APE may be introduced as a possible anti-aging agent.


Subject(s)
Andrographis/chemistry , Endothelial Progenitor Cells/drug effects , Plant Extracts/therapeutic use , Skin Aging/drug effects , Adult , Animals , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Elasticity/drug effects , Endothelial Progenitor Cells/physiology , Female , Gene Expression Regulation/drug effects , Humans , Integrin beta1/metabolism , Mice , Middle Aged , Plant Extracts/administration & dosage , Plant Extracts/chemistry
9.
J Rehabil Med ; 47(1): 87-93, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25224020

ABSTRACT

OBJECTIVE: To locate and trace endogenous endothelial progenitor cells (EPCs) in rabbits subjected to myocardial ischaemia and/or physiological ischaemia training. METHODS: Rabbits were randomly divided into 4 groups: a myocardial ischaemia group (subjected to myocardial ischaemia only); a physiological ischaemia training group (subjected to physiological ischaemia training only); a physiological ischaemia training-myocardial ischaemia group (subjected to both myocardial ischaemia and physiological ischaemia training); and a sham-operated group. Myocardial ischaemia was induced experimentally by a 2-min ischaemia, followed by a 1-h reperfusion. Physiological ischaemia training involved a 4-min isometric contraction elicited by electrical stimulation (biphase square wave, 40 Hz, 1 ms), which generated a contraction force at 40% of the maximal isometric contraction force. Myocardial ischaemia I and/or physiological ischaemia training were performed twice a day, 5 days a week for 4 weeks. Capillary densities and EPC levels in both blood and the ischaemic heart region were then measured. EPCs were traced by double-labelling with super paramagnetic iron oxide and chloromethyl-benzamidodialkylcarbocyanine. RESULTS: EPC levels in the blood and the ischaemic heart region both improved significantly in the physiological ischaemia training-myocardial ischaemia group (mean 0.046% (standard deviation (SD) 0.007), 0.013% (SD 0.005)) and group myocardial ischaemia (mean 0.038% (SD 0.016), 0.008% (SD 0.004)). For the physiological ischaemia training group, moderately raised EPCs were found in the blood (0.026 ± 0.010%), but not in the heart. Capillary density increased in the physiological ischaemia training-myocardial ischaemia and myocardial ischaemia groups. The dual-labelled EPCs were confirmed in the ischaemic heart region. Pearson's analysis demonstrated that there is a positive correlation between EPC levels in the blood and the heart region (p < 0.05), and between circulating EPCs and the capillary (p < 0.05) for the physiological ischaemia training-myocardial ischaemia group. CONCLUSION: Physiological ischaemia training can effectively improve endogenous EPCs. Their homing process from the circulating blood to the ischaemic myocardium was clearly traced in this study on rabbits. This homing process is of great importance for remote neovascularization.


Subject(s)
Electric Stimulation Therapy/methods , Endothelial Progenitor Cells/physiology , Myocardial Ischemia/pathology , Myocardial Ischemia/therapy , Animals , Capillaries/pathology , Cell Count , Cell Movement/physiology , Coronary Circulation/physiology , Endothelial Progenitor Cells/transplantation , Isometric Contraction/physiology , Male , Neovascularization, Physiologic/physiology , Rabbits
10.
Atherosclerosis ; 236(2): 400-10, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25150938

ABSTRACT

OBJECTIVE: Obstructive sleep apnoea (OSA) has been implicated as a risk factor for atherosclerosis. The aim of our study was to examine the effects of chronic intermittent hypoxia in apoE-/- mice serving as model of OSA on endothelial dysfunction and oxidative stress and to evaluate the reversibility of hypoxia-induced changes under anti-inflammatory infliximab and anti-oxidative l-glutathione. METHODS: ApoE-/- mice were divided into 4 groups (n = 9 each): 1. intermittent hypoxia 8 h/day for 6 weeks, 2. intermittent hypoxia + injections of infliximab, 3. intermittent hypoxia + injections of l-glutathione, 4. normoxia = control. RESULTS: Endothelial function was impaired under hypoxia compared to control. Application of infliximab and l-glutathione improved it to a level of control. The percentage of endothelial microparticles increased under hypoxia compared to other groups. Levels of NADPH oxidase 2-derived reactive oxygen species were approximately 9 times higher in the hypoxia group. The number of sca-1/flk-1+ endothelial progenitor cells was higher in bone marrow and lower in blood under hypoxia vs. other groups. Stromal cell derived factor-1alpha- and matrix metalloproteinase-9-dependent release of these cells from bone marrow was attenuated under hypoxia. The number of DilacLDL+/lectin + early outgrowth progenitor cells and that of colony forming units from these cells were higher under hypoxia. Atherosclerotic plaques in the aorta were more frequent under hypoxia and control in comparison with both drug groups. CONCLUSION: Intermittent hypoxia contributes to endothelial dysfunction by the local increase in reactive oxygen species and reduction of the peripheral repair capacity. Infliximab and l-glutathione prevent hypoxia-induced vascular and extravascular changes.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antioxidants/therapeutic use , Endothelium, Vascular/physiopathology , Glutathione/therapeutic use , Hypoxia/physiopathology , Animals , Aortic Diseases/etiology , Apolipoproteins E/deficiency , Bone Marrow/physiopathology , Cell-Derived Microparticles , Cells, Cultured , Disease Models, Animal , Drug Evaluation, Preclinical , Endothelial Progenitor Cells/physiology , Female , Hypoxia/blood , Hypoxia/drug therapy , Infliximab , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress , Plaque, Atherosclerotic/etiology , Reactive Oxygen Species/blood , Sleep Apnea, Obstructive , Spleen/cytology
11.
Chin Med J (Engl) ; 127(11): 2121-8, 2014.
Article in English | MEDLINE | ID: mdl-24890165

ABSTRACT

BACKGROUND: Diabetes mellitus (DM) is a common disease accompanied with a high incidence of hind limb ischemia (HLI). In recent years, numerous studies demonstrated that endothelial progenitor cells (EPCs) are involved in angiogenesis and maintenance of vascular integrity following HLI. On the other side, it has been proved that Astragalus polysaccharide (APS) could promote angiogenesis. In the present study, we aimed to evaluate the effect of APS and EPCs on enhancing angiogenesis after experimental HLI caused by femoral artery ligation in rats with streptozotocin (STZ)-induced diabetes. METHODS: Rats (n = 110) were randomly assigned to the following groups: sham group, ischemia group, APS group, EPCs group and APS+EPCs group. APS, EPCs or an equal volume of vehicle was administered intramuscularly after HLI induction, and 6 rats were assessed by angiography at 28 days after induction of HLI, 6 rats were sacrificed at the same time point to take histological studies, biochemical tests were also performed at that point in the rest rats. RESULTS: APS or EPCs treatment induced an increase, respectively, in the protein expression of vascular endothelial growth factor (VEGF) (36.61%, 61.59%), VEGF receptor-1 (VEGFR-1) (35.50%, 57.33%), VEGFR-2 (31.75%, 41.89%), Angiopoietin-1 (Ang-1) (37.57%, 64.66%) and Tie-2 (42.55%, 76.94%) (P < 0.05), after HLI injury. And combined therapy of APS and EPCs enhanced the effort of angiogenesis after HLI induction in diabetic rats, through elevating protein expression of VEGF (99.67%), VEGFR-1 (105.33%), VEGFR2 (72.05%), Ang-1 (114.30%) and Tie-2 (111.87%) (P < 0.05). Similarly, mRNA expression of VEGF, VEGFR-1, VEGFR2, Ang-1, Tie-2 also show similar trends as well as protein expression (P < 0.05). CONCLUSION: APS or EPCs could enhance angiogenesis, and the combined treatment leads to better effort, at least, partially via VEGF/VEGFR and Ang-1/Tie-2 signaling pathway.


Subject(s)
Astragalus Plant/chemistry , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/therapy , Endothelial Progenitor Cells/physiology , Ischemia/drug therapy , Ischemia/therapy , Polysaccharides/therapeutic use , Animals , Hindlimb/pathology , Male , Rats
SELECTION OF CITATIONS
SEARCH DETAIL