Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
2.
Int J Mol Sci ; 22(9)2021 Apr 29.
Article in English | MEDLINE | ID: mdl-33946902

ABSTRACT

Previous work showed a link between Tie2+ nucleus pulposus progenitor cells (NPPC) and disc degeneration. However, NPPC remain difficult to maintain in culture. Here, we report whole tissue culture (WTC) combined with fibroblast growth factor 2 (FGF2) and chimeric FGF (cFGF) supplementation to support and enhance NPPC and Tie2 expression. We also examined the role of PI3K/Akt and MEK/ERK pathways in FGF2 and cFGF-induced Tie2 expression. Young herniating nucleus pulposus tissue was used. We compared WTC and standard primary cell culture, with or without 10 ng/mL FGF2. PI3K/Akt and MEK/ERK signaling pathways were examined through western blotting. Using WTC and primary cell culture, Tie2 positivity rates were 7.0 ± 2.6% and 1.9 ± 0.3% (p = 0.004), respectively. Addition of FGF2 in WTC increased Tie2 positivity rates to 14.2 ± 5.4% (p = 0.01). FGF2-stimulated expression of Tie2 was reduced 3-fold with the addition of the MEK inhibitor PD98059 (p = 0.01). However, the addition of 1 µM Akt inhibitor, 124015-1MGCN, only reduced small Tie2 expression (p = 0.42). cFGF similarly increased the Tie2 expression, but did not result in significant phosphorylation in both the MEK/ERK and PI3K/Akt pathways. WTC with FGF2 addition significantly increased Tie2 maintenance of human NPPC. Moreover, FGF2 supports Tie2 expression via MEK/ERK and PI3K/Akt signals. These findings offer promising tools and insights for the development of NPPC-based therapeutics.


Subject(s)
Fibroblast Growth Factor 2/pharmacology , Nucleus Pulposus/drug effects , Receptor, TIE-2/biosynthesis , Signal Transduction/drug effects , Adolescent , Adult , Cells, Cultured , Collagen Type II/biosynthesis , Collagen Type II/genetics , Female , Fibroblast Growth Factor 1/genetics , Fibroblast Growth Factor 2/genetics , Flavonoids/pharmacology , Humans , Intervertebral Disc Displacement/pathology , MAP Kinase Signaling System/drug effects , Male , Nucleus Pulposus/cytology , Nucleus Pulposus/metabolism , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/physiology , Receptor, TIE-2/genetics , Recombinant Fusion Proteins/pharmacology , Stem Cells/drug effects , Stem Cells/metabolism , Young Adult
3.
Exp Eye Res ; 193: 107972, 2020 04.
Article in English | MEDLINE | ID: mdl-32059975

ABSTRACT

Autophagy plays critical roles in various ocular diseases, including age-related macular degeneration (AMD). Tie2-expressing macrophages (TEMs) play crucial roles in angiogenesis. To investigate the role of TEMs and autophagy in the development of AMD, we employed macrophage-specific Tie2 knockout mice and used a laser-induced choroidal neovascularization (CNV). The results showed that TEMs can promote CNV formation by up-regulating the level of autophagy. These results were further verified by in vitro cell experiments that peritoneal macrophages from Tie2 knockout mice can inhibit the expression of autophagy-related factors and inhibit the expression of angiogenic factor of VEGF by activating AMPK signaling pathway. Our results suggest that TEMs and macrophage Tie2 signal mediated-autophagy play critical role in experimental CNV, and they may be novel preventive targets for AMD treatment.


Subject(s)
Choroidal Neovascularization/genetics , Gene Expression Regulation , Macrophages/pathology , Receptor, TIE-2/genetics , Animals , Autophagy , Cells, Cultured , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Disease Models, Animal , Lasers/adverse effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, TIE-2/biosynthesis , Signal Transduction
4.
Spine (Phila Pa 1976) ; 44(23): 1613-1622, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31730570

ABSTRACT

STUDY DESIGN: Experimental study with human mesenchymal stem cells (MSCs) and intervertebral disc (IVD) tissue samples. OBJECTIVE: This study aimed to characterize the effect of MSC homing on the Tie2-positive IVD progenitor cell population, IVD cell survival, and proliferation. SUMMARY OF BACKGROUND DATA: Homing of human MSCs has been described as potential alternative to MSC injection, aiming to enhance the regenerative capacity of the IVD. IVD cells expressing Tie2 (also known as CD202b or Angiopoietin-1 receptor TEK tyrosine kinase) represent a progenitor cell population with discogenic differentiation potential. However, the fraction of Tie2-positive progenitor cells decreases with aging and degree of IVD degeneration, resulting in a potential loss of the IVD's regenerative capacity. METHODS: Human MSCs, isolated from vertebral bone marrow aspirates, were labeled and seeded onto the endplate of bovine IVDs and human IVD tissue. Following MSC migration for 5 days, IVD cells were isolated by tissue digestion. The fractions of Tie2-positive, dead, apoptotic, and proliferative IVD cells were evaluated by flow cytometry and compared to untreated IVDs. For human IVDs, 3 groups were investigated: nondegenerated (organ donors), IVDs of patients suffering from spinal trauma, and degenerative IVD tissue samples. RESULTS: MSC homing enhanced the fraction of Tie2-positive IVD cells in bovine and human IVD samples. Furthermore, a proliferative response and lower fraction of dead cells were observed after MSC homing in both bovine and human IVD tissues. CONCLUSION: Our findings indicate that MSC homing enhances the survival and regenerative capability of IVD cells, which may be mediated by intercellular communication. MSC homing could represent a potential treatment strategy to prevent the onset of the degenerative cascade in IVDs at risk such as IVDs adjacent to a fused segment or IVDs after herniation. LEVEL OF EVIDENCE: N/A.


Subject(s)
Cell Proliferation/physiology , Intervertebral Disc/metabolism , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Receptor, TIE-2/biosynthesis , Animals , Cattle , Cell Death/physiology , Cell Differentiation/physiology , Cells, Cultured , Female , Humans , Intervertebral Disc/pathology , Intervertebral Disc Degeneration/metabolism , Intervertebral Disc Degeneration/therapy , Organ Culture Techniques
5.
Br J Anaesth ; 121(5): 1041-1051, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30336848

ABSTRACT

BACKGROUND: Cardiopulmonary bypass (CPB) during cardiac surgery impairs microcirculatory perfusion and is paralleled by vascular leakage. The endothelial angiopoietin/Tie2 system controls microvascular leakage. This study investigated whether targeting Tie2 with the angiopoietin-1 mimetic vasculotide reduces vascular leakage and preserves microcirculatory perfusion in a rat CPB model. METHODS: Rats were subjected to 75 min of CPB after treatment with vasculotide or phosphate buffered solution as control or underwent a sham procedure. Microcirculatory perfusion and leakage were assessed with intravital microscopy (n=10 per group) and Evans blue dye extravasation (n=13 per group), respectively. Angiopoietin-1, -2, and Tie2 protein and gene expression were determined in plasma, kidney, and lung. RESULTS: CPB immediately impaired microcirculatory perfusion [5 (4-8) vs 10 (7-12) vessels per recording, P=0.002] in untreated CPB rats compared with sham, which persisted after weaning from CPB. CPB increased circulating angiopoeietin-1, -2, and soluble Tie2 concentrations and reduced Tie2 messenger ribonucleic acid (mRNA) expression in kidney and lung. Moreover, CPB increased Evans blue dye leakage in kidney [12 (8-25) vs 7 (1-12) µg g-1, P=0.04] and lung [and 23 (13-60) vs 6 (4-16) µg g-1, P=0.001] compared with sham. Vasculotide treatment preserved microcirculatory perfusion during and after CPB. Moreover, vasculotide treatment reduced Evans blue dye extravasation in lung compared with CPB control [18 (6-28) µg g-1vs 23 (13-60) µg g-1, P=0.04], but not in kidney [10 (3-23) vs 12 (8-25) µg g-1, P=0.38]. Vasculotide did not affect circulating or mRNA expression of angiopoietin-1, -2, and Tie2 concentrations compared with untreated CPB controls. CONCLUSIONS: Treatment with the angiopoietin-1 mimetic vasculotide reduced pulmonary vascular leakage and preserved microcirculatory perfusion during CPB in a rat model.


Subject(s)
Angiopoietin-1/therapeutic use , Cardiopulmonary Bypass/adverse effects , Peptide Fragments/therapeutic use , Pulmonary Circulation/drug effects , Angiopoietin-1/biosynthesis , Angiopoietin-1/genetics , Angiopoietin-2/biosynthesis , Angiopoietin-2/genetics , Animals , Capillaries/drug effects , Gene Expression/drug effects , Male , Microcirculation/drug effects , Rats , Rats, Wistar , Receptor, TIE-2/biosynthesis , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism
6.
Crit Care Med ; 46(9): e928-e936, 2018 09.
Article in English | MEDLINE | ID: mdl-29979219

ABSTRACT

OBJECTIVES: Tie2 is a tyrosine kinase receptor expressed by endothelial cells that maintains vascular barrier function. We recently reported that diverse critical illnesses acutely decrease Tie2 expression and that experimental Tie2 reduction suffices to recapitulate cardinal features of the septic vasculature. Here we investigated molecular mechanisms driving Tie2 suppression in settings of critical illness. DESIGN: Laboratory and animal research, postmortem kidney biopsies from acute kidney injury patients and serum from septic shock patients. SETTING: Research laboratories and ICU of Hannover Medical School, Harvard Medical School, and University of Groningen. PATIENTS: Deceased septic acute kidney injury patients (n = 16) and controls (n = 12) and septic shock patients (n = 57) and controls (n = 22). INTERVENTIONS: Molecular biology assays (Western blot, quantitative polymerase chain reaction) + in vitro models of flow and transendothelial electrical resistance experiments in human umbilical vein endothelial cells; murine cecal ligation and puncture and lipopolysaccharide administration. MEASUREMENTS AND MAIN RESULTS: We observed rapid reduction of both Tie2 messenger RNA and protein in mice following cecal ligation and puncture. In cultured endothelial cells exposed to tumor necrosis factor-α, suppression of Tie2 protein was more severe than Tie2 messenger RNA, suggesting distinct regulatory mechanisms. Evidence of protein-level regulation was found in tumor necrosis factor-α-treated endothelial cells, septic mice, and septic humans, all three of which displayed elevation of the soluble N-terminal fragment of Tie2. The matrix metalloprotease 14 was both necessary and sufficient for N-terminal Tie2 shedding. Since clinical settings of Tie2 suppression are often characterized by shock, we next investigated the effects of laminar flow on Tie2 expression. Compared with absence of flow, laminar flow induced both Tie2 messenger RNA and the expression of GATA binding protein 3. Conversely, septic lungs exhibited reduced GATA binding protein 3, and knockdown of GATA binding protein 3 in flow-exposed endothelial cells reduced Tie2 messenger RNA. Postmortem tissue from septic patients showed a trend toward reduced GATA binding protein 3 expression that was associated with Tie2 messenger RNA levels (p < 0.005). CONCLUSIONS: Tie2 suppression is a pivotal event in sepsis that may be regulated both by matrix metalloprotease 14-driven Tie2 protein cleavage and GATA binding protein 3-driven flow regulation of Tie2 transcript.


Subject(s)
Receptor, TIE-2/physiology , Sepsis/physiopathology , Adult , Aged , Animals , Cells, Cultured , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Prospective Studies , Receptor, TIE-2/biosynthesis
7.
Curr Eye Res ; 43(9): 1190-1198, 2018 09.
Article in English | MEDLINE | ID: mdl-29857790

ABSTRACT

PURPOSE: To study retinal neovascularization (RNV) inhibition by intravitreal injections (IVs) of ranibizumab, sTie2 fusion protein (sTie2-Fc), and a combined therapy in an oxygen-induced retinopathy (OIR) model. MATERIALS AND METHODS: An OIR mouse model was used to simulate RNV in retinopathy of prematurity (ROP); and the effect of blocking the angiopoietin (Ang) and its receptor (Tie2) and the vascular endothelial growth factor (VEGF) and its receptor (VEGFR) signaling pathways was compared using an IV of sTie2-Fc (Ang inhibitor) and/or ranibizumab (aVEGF antagonist). The effects were assessed using fluorescein isothiocyanate (FITC)-dextran cardiac perfusion, isolectin B4 (IB4) staining with whole retinal mounting, and hematoxylin and eosin (HE) staining to count the endothelial cells (ECs) that broke through the internal limiting membrane (ILM). The mRNA and protein levels of VEGF-A, VEGFR-2, Ang1, Ang2, and Tie2 were also determined by reverse transcriptase polymerase chain reaction (RT-PCR) and western blot analysis. RESULTS: Compared with the control group injected with phosphate-buffered saline (PBS), all three experimental groups, ranibizumab, sTie2-Fc, and ranibizumab + sTie2-Fc, had a significant decrease in micro-vessel densities and neovascular clusters, and fewer ECs broke through the ILM (all p < 0.05). The non-perfusion areas decreased in both mono-treated groups, although the combined therapy had larger non-perfusion areas. All three treatments decreased the mRNA and protein levels of VEGFA, Ang1, and Tie2. CONCLUSION: In this study, it was confirmed that blocking the Ang/Tie2 and/or VEGF/VEGFR pathways could inhibit RNV and decrease abnormal micro-vessel density; and the mono-blockage of Ang/Tie2 might cause a smaller non-perfusion area.


Subject(s)
Gene Expression Regulation, Developmental , Ranibizumab/administration & dosage , Receptor, TIE-2/genetics , Retina/metabolism , Retinal Neovascularization/drug therapy , Angiogenesis Inhibitors/administration & dosage , Animals , Animals, Newborn , Blotting, Western , Disease Models, Animal , Intravitreal Injections , Mice , Mice, Inbred C57BL , Oxygen/toxicity , RNA/genetics , Receptor, TIE-2/biosynthesis , Retina/drug effects , Retina/pathology , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Vascular Endothelial Growth Factor A/antagonists & inhibitors
8.
Tumour Biol ; 39(7): 1010428317711658, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28720059

ABSTRACT

Metastatic or recurrent cervical cancer has limited treatment options and a high rate of mortality. Although anti-vascular endothelial growth factor drugs have shown great promise as a therapeutic target for treatment of advanced cervical cancer, drug resistance and class-specific side effects negate long-term benefits. The identification of alternative anti-angiogenic factors will be critical for future drug development for advanced or recurrent cervical cancer. In this study, we found that angiopoietins and Tie receptors were highly expressed in cervical cancer cells. Tie-2 expression in tumor cells predicted poorer prognosis. Wound closure assay and Transwell assay showed that upregulated or downregulated Ang-1 and Ang-2 expression promoted or reduced cervical cancer cell lines migration and invasion, respectively. In subcutaneous xenograft models of cervical cancer, downregulation of Ang-1 and Ang-2 attenuated tumor growth. The expression of vimentin and endomucin and microvessel density were all significantly decreased in the siAng-1 group and siAng-2 group relative to the infection control group. Our data support that dual inhibition of Ang-1 and Ang-2 may be an alternative target for anti-angiogenic adjuvant therapy in advanced or recurrent cervical squamous cell cancer.


Subject(s)
Angiopoietin-1/genetics , Receptor, TIE-2/genetics , Uterine Cervical Neoplasms/genetics , Vesicular Transport Proteins/genetics , Adult , Aged , Angiopoietin-1/biosynthesis , Animals , Carcinogenesis/genetics , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Mice , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Prognosis , Receptor, TIE-2/biosynthesis , Uterine Cervical Neoplasms/pathology , Vesicular Transport Proteins/biosynthesis , Xenograft Model Antitumor Assays
9.
Int J Cancer ; 141(5): 1011-1017, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28555943

ABSTRACT

Sorafenib, a multi-kinase inhibitor, inhibits tumor angiogenesis and is the first-line systemic therapy for patients with advanced hepatocellular carcinoma (HCC). However, due to its limited effects and frequent occurrence of side effects, biomarkers are needed to predict the effects of sorafenib. We considered the possibility of using TIE-2-expressing monocytes (TEMs) to predict the response in sorafenib-treated patients with advanced HCC. TEMs serve as a diagnostic marker of HCC and are related to angiogenesis. We analyzed 25 advanced HCC patients and prospectively evaluated TEMs before (Pre TEMs) and at 1 month after initial therapy (T1m TEMs). The radiologic response was evaluated by modified Response Evaluation Criteria in Solid Tumors (mRECIST). Median survival time (MST) was significantly longer in the partial response/stable disease (PR/SD) group (21.8 months) than in the PD group (8.7 months). ΔTEMs (changes of T1m TEMs compared to Pre TEMs) were significantly lower in the PR/SD group than in the PD group. MST of the ΔTEMs low group (14.2 months) was significantly longer than that of the high group (8.7 months). Univariate and multivariate Cox regression analyses showed that ΔTEMs [hazard ratio (HR) = 8.53, 95% confidence interval (CI) = 1.51-48.16, p = 0.015] and Child-Pugh class (HR = 5.59, 95% CI = 1.06-29.63, p = 0.043) were independently associated with overall survival. Our results suggest that ΔTEMs could serve as a biomarker for predicting radiologic response and overall survival in sorafenib-treated patients with advanced HCC.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/blood , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Receptor, TIE-2/biosynthesis , Aged , Aged, 80 and over , Angiogenesis Inhibitors/therapeutic use , Area Under Curve , Female , Flow Cytometry , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Monocytes , Niacinamide/therapeutic use , Pilot Projects , Proportional Hazards Models , Prospective Studies , ROC Curve , Response Evaluation Criteria in Solid Tumors , Sorafenib
10.
PLoS One ; 12(4): e0174555, 2017.
Article in English | MEDLINE | ID: mdl-28369068

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is the second common cancer in Henan province and is well-known for aggressiveness and dismal prognosis. Adjuvant therapies, chemotherapy, radiotherapy and endoscopic treatment have not improved survival rates in patients with late stage esophageal carcinoma. All-trans retinoic acid (ATRA) is the active ingredient of Vitamin A and affects a wide spectrum of biological processes including development, growth, neural function, immune function, reproduction, and vision. It is one of the most potent therapeutic agents used for treating cancers, especially lung adenocarcinomas. ATRA inhibits metastatic potential and angiogenesis in several tumor models. We investigated the effects of ATRA on the expression of angiopoietin 1 (Ang-1), angiopoietin 2 (Ang-2) and receptor Tie-2 in EC1 cells in vitro. We also assessed the growth and migration of EC1 cells in vitro. ATRA treatment caused 29.5% and 40.3% reduction of the growth of EC1 cells after 24 hours and 48 hours, relative to the control. ATRA plus fluorouracil treatment reduced the viability more strongly than either drug alone, indicating an additive effect. Moreover, ATRA decreased EC1 migration by 87%. Furthermore, ATRA treatment led to a marked decrease of the transcript levels of Ang-1, Ang-2, Tie-2, VEGF, and VEGF receptors, as assessed by real-time RT-PCR. Importantly, the protein levels of Ang-1, Ang-2 and Tie-2 were reduced by ATRA treatment. In vivo, we found ATRA treatment suppressed the tumor growth and improved the cachexia of mice. Importantly, ATRA treatment decreased the expression of CD31, Ang-1, Ang-2 and Tie-2 in subcutaneous tumors of EC1 cells. Collectively, our findings demonstrate that ATRA exhibits a dose- and temporal-dependent effect on the metastatic behavior, suppresses the angiopoietin-Tie2 pathway and inhibits angiogenesis and the progression of xenograft tumors of EC1 cells.


Subject(s)
Angiopoietin-1/biosynthesis , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Esophageal Neoplasms/drug therapy , Neoplasm Metastasis/drug therapy , Neovascularization, Pathologic/drug therapy , Receptor, TIE-2/biosynthesis , Tretinoin/pharmacology , Vesicular Transport Proteins/biosynthesis , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Esophageal Squamous Cell Carcinoma , Female , Fluorouracil/pharmacology , Humans , Mice , Mice, Inbred BALB C , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Receptors, Vascular Endothelial Growth Factor/biosynthesis , Vascular Endothelial Growth Factor A/biosynthesis
11.
Neurosci Lett ; 644: 62-66, 2017 03 22.
Article in English | MEDLINE | ID: mdl-28219791

ABSTRACT

Our previous studies have found that stem cells conditioned medium (CM) facilitated functional recovery after stroke in non-diabetics. However, whether bone marrow stromal cells conditioned medium (BMSCs-CM) treatment after stroke in type 2 diabetic (T2DM) rats improve functional outcomes remains unclear. T2DM rats were induced and subjected to stroke then treated with or without BMSCs-CM. Functional outcomes and blood-brain barrier (BBB) leakage were performed, the expression of Angiopoietin (Ang) 1 and tyrosine kinase (Tie) 2 were also assessed. Our results showed that BMSCs-CM treatment significantly improved functional outcomes, decreased BBB leakage and the expression of Ang1 and Tie2 were also changed after BMSCs-CM treatment in type 2 diabetes after stroke. In conclusion, enhanced expression of Ang1 and Tie2 in ischemic brain after BMSCs-CM treatment of stroke may contribute to the improved functional recovery after stroke in type 2 diabetic rats.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Mesenchymal Stem Cells/drug effects , Recovery of Function , Stroke/pathology , Vascular Remodeling , Angiopoietin-1/biosynthesis , Animals , Bone Marrow Cells , Brain/metabolism , Brain/pathology , Culture Media, Conditioned/pharmacology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2 , Male , Mesenchymal Stem Cell Transplantation , Rats , Rats, Wistar , Receptor, TIE-2/biosynthesis , Recovery of Function/drug effects , Stroke/complications , Stroke/metabolism
12.
Clin Exp Pharmacol Physiol ; 44(1): 123-131, 2017 01.
Article in English | MEDLINE | ID: mdl-27712004

ABSTRACT

Increased pulmonary vascular resistance is a critical complication in sepsis. Toll-like receptor (TLR) as well as angiopoietin (ANG) signalling both contribute to the emergence of pulmonary arterial hypertension. We hypothesized that TLR stimulation by bacterial ligands directly affects expression and secretion of ligands and receptors of the angiopoietin/TIE axis. Microvascular endothelial (HPMEC) and smooth muscle cells (SMC) of pulmonary origin were incubated with thrombin and with ligands for TLR2, -4, -5, and -9. Expression and secretion of ANG1, -2, TIE2 and IL-8 were determined using quantitative real-time PCR and ELISA. TLR stimulation had no impact either on the expression of ANG2 and TIE2 in HPMEC or on that of ANG1 in SMC. However, overall levels of both released ANG1 and -2 were halved upon stimulation with the TLR9 ligand CpG, and ANG2 release was significantly enhanced by TLR4 activation when initially provoked by sequentially performed stimulation. Furthermore, enhanced ANG2 activity increased endothelial permeability, as demonstrated in an in vitro transwell assay. We conclude that sole TLR stimulation by bacterial ligands plays no significant role for altered expression and secretion of ANG1, -2 and TIE2 in human pulmonary vascular cells. The interplay between various stimuli is required to induce imbalances between ANG1 and -2.


Subject(s)
Angiopoietin-1/biosynthesis , Angiopoietin-2/biosynthesis , Pulmonary Artery/metabolism , Receptor, TIE-2/biosynthesis , Toll-Like Receptors/biosynthesis , Angiopoietins/biosynthesis , Cells, Cultured , Flagellin/toxicity , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation Mediators/metabolism , Microvessels/drug effects , Microvessels/metabolism , Pneumonia/chemically induced , Pneumonia/metabolism , Pulmonary Artery/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology
13.
PLoS One ; 11(7): e0159532, 2016.
Article in English | MEDLINE | ID: mdl-27439004

ABSTRACT

We have recently reported that Compound 49b, a novel ß-adrenergic receptor agonist, can significantly reduce VEGF levels in retinal endothelial cells (REC) grown in diabetic-like conditions. In this study, we investigated whether Compound 49b could protect the retina under hypoxic conditions using the ischemia-reperfusion (I/R)-induced model in rats, as well REC cultured in hypoxic conditions. Some rats received 1mM topical Compound 49b for the 2 (5 rats each group) or 10 (4 rats in each group) days post-I/R. Analyses for retinal thickness and cell loss in the ganglion cell layer was done at 2 days post-I/R, while numbers of degenerate capillaries and pericyte ghosts were measured at 10 days post-I/R. Additionally, REC were cultured in normal oxygen or hypoxia (5% O2) only or treated with 50 nM Compound 49b for 12 hours. Twelve hours after Compound 49b exposure, cells were collected and analyzed for protein levels of insulin-like growth factor binding protein 3 (IGFBP-3), vascular endothelial cell growth factor (VEGF) and its receptor (KDR), angiopoietin 1 and its receptor Tie2 for Western blotting. Data indicate that exposure to I/R significantly decreased retinal thickness, with increasing numbers of degenerate capillaries and pericyte ghosts. Compound 49b treatment inhibited these retinal changes. In REC cultured in hypoxia, levels of IGFBP-3 were reduced, which were significantly increased by Compound 49b. Hypoxia significantly increased protein levels of VEGF, KDR, Angiopoiein 1, and Tie2, which were reduced following Compound 49b treatment. These data strongly suggested that Compound 49b protected the retina against I/R-induced injury. This provides additional support for a role of ß-adrenergic receptor actions in the retina.


Subject(s)
Adrenergic beta-Agonists/administration & dosage , Angiopoietin-1/biosynthesis , Diabetic Retinopathy/drug therapy , Insulin-Like Growth Factor Binding Protein 3/biosynthesis , Receptor, TIE-2/biosynthesis , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor Receptor-2/biosynthesis , Angiopoietin-1/genetics , Animals , Apoptosis/drug effects , Cell Hypoxia/genetics , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Diabetic Retinopathy/genetics , Diabetic Retinopathy/pathology , Endothelial Cells/drug effects , Endothelial Cells/pathology , Gene Expression Regulation/drug effects , Humans , Insulin-Like Growth Factor Binding Protein 3/genetics , Phosphorylation , Rats , Receptor, TIE-2/genetics , Reperfusion Injury/drug therapy , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Retina/drug effects , Retina/pathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
14.
Gene ; 580(1): 1-7, 2016 Apr 10.
Article in English | MEDLINE | ID: mdl-26748243

ABSTRACT

Tie2-expressing Monocytes (TEMs) were previously identified as a novel subset of monocytes and were believed to have prominent pro-angiogenesis activities in human tumors. While the molecular mechanism of the angiogenesis promoting capacity of TEMs remains unclear. RNA transcriptome pattern, including non-coding RNAs as microRNA (miRNA) and long non-coding RNA (lncRNA), plays important role in cell differentiation and functions. However, little is known about the transcriptome patterns of TEMs, including those non-coding RNAs. We explore the transcriptome of TEMs and the matched monocytes that do not express Tie2 (Tie2(-)monocytes) isolated from peripheral blood of healthy adults employing the Agilent Human miRNA(8*60K,Design ID: 046064)microarray and the Agilent lncRNA Gene Expression(4*180K, Design ID: 042818)microarray. A total of 141 mRNAs, 142 lncRNAs and 75 miRNAs were found dysregulated in TEMs compared to Tie2(-)monocytes. TEMs have the distinct RNA transcriptome patterns according to the Hierarchical clustering and then the gene expression patterns were confirmed by quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR). Functional annotation by Gene Ontology (GO) analyses showed that the up-regulated mRNAs in TEMs were associated to blood vessel remodeling and positive regulation of epithelial cell proliferation, and the up-regulated insulin like growth factor 1(IGF1) mRNA was involved in both pathways. For functional analysis of those dysregulated non-coding RNAs, target genes of the miRNAs were predicted and cis/trans-regulation analysis of the lncRNAs were performed.


Subject(s)
Leukocytes, Mononuclear/metabolism , MicroRNAs/genetics , Neovascularization, Pathologic/genetics , RNA, Long Noncoding/genetics , Receptor, TIE-2/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , Epithelial Cells/cytology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Insulin-Like Growth Factor I/biosynthesis , Insulin-Like Growth Factor I/genetics , Leukocytes, Mononuclear/cytology , MicroRNAs/biosynthesis , Neoplasms/blood supply , Neoplasms/genetics , Oligonucleotide Array Sequence Analysis , RNA, Long Noncoding/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, TIE-2/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Transcriptome/genetics
15.
J Sex Med ; 12(4): 876-86, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25487286

ABSTRACT

INTRODUCTION: Erectile dysfunction is highly prevalent in patients with advanced age or cardiovascular disease risk factors (CVDRFs). These conditions interfere on expression of vascular growth factors and respective receptors causing disturbance in endothelial function. AIM: This study aims to assess the effect of aging and CVDRF on the expression of tyrosine kinase with immunoglobulin-like and EGF-like domains (Tie) 1 in human corpus cavernosum (CC). METHODS: CC fragments obtained from programmed surgeries or organ donors were divided into three groups: young, healthy aged, and aged with CVDRF. Angiopoietin (Ang) 1, Ang2, Tie1, and Tie2 mRNA and protein levels were assessed by real-time polymerase chain reaction and Western blotting, respectively. Dual-immunolabeling of Tie1 with specific markers of endothelium and smooth muscle and Ang1 and Ang2 was performed. MAIN OUTCOME MEASURES: To characterize the expression of Tie1 in human CC and elucidate its potential inhibitory effect in Ang-Tie2 system. RESULTS: Analysis of mRNAs demonstrated a decrease in Tie1 expression in CVDRF individuals compared with aged or young healthy individuals. No variation for Tie2, Ang1, or Ang2 expression was observed among the studied groups. In all analyzed CC fragments, a 125 kDa band, Tie1, was detected. This protein presented a significant age-related decrease, specially in individuals with CVDRF. Immunofluorescence study revealed Tie1 expression in the endothelium of samples of all experimental groups. CONCLUSIONS: Employing different methodological approaches, we show for the first time that Tie1 is expressed in human CC endothelium, and its level of expression diminishes in aged individuals, particularly those with CVDRF. This finding reinforces the view that delivery of Ang1 to the CC of erectile dysfunction affected CVDRF patients is able to activate a beneficial Tie2 response.


Subject(s)
Aging/metabolism , Cardiovascular Diseases/physiopathology , Penis/metabolism , Receptor, TIE-1/biosynthesis , Adult , Age Factors , Aged , Angiopoietin-1/biosynthesis , Angiopoietin-2/biosynthesis , Blotting, Western , Endothelium/metabolism , Gene Expression , Humans , Male , Middle Aged , Polymerase Chain Reaction , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor, TIE-2/biosynthesis , Risk Factors
16.
Int J Oncol ; 45(5): 2058-64, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25176420

ABSTRACT

The α6 integrin subunit (α6) has been implicated in cancer cell migration and in the progression of several malignancies, but its role in tumor angiogenesis is unclear. In mice, anti-α6 blocking antibodies reduce tumor angiogenesis, whereas Tie1-dependent α6 gene deletion enhances neovessel formation in melanoma and lung carcinoma. To clarify the discrepancy in these results we used the cre-lox system to generate a mouse line, α6fl/fl­Tie2Cre(+), with α6 gene deletion specifically in Tie2-lineage cells: endothelial cells, pericytes, subsets of hematopoietic stem cells, and Tie2-expressing monocytes/macrophages (TEMs), known for their proangiogenic properties. Loss of α6 expression in α6fl/fl­Tie2Cre(+) mice reduced tumor growth in a murine B16F10 melanoma model. Immunohistological analysis of the tumors showed that Tie2-dependent α6 gene deletion was associated with reduced tumor vascularization and with reduced infiltration of proangiogenic Tie2-expressing macrophages. These findings demonstrate that α6 integrin subunit plays a major role in tumor angiogenesis and TEM infiltration. Targeting α6 could be used as a strategy to reduce tumor growth.


Subject(s)
Integrin alpha6/genetics , Melanoma, Experimental/genetics , Neovascularization, Pathologic/genetics , Receptor, TIE-2/genetics , Animals , Cell Lineage , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Humans , Integrin alpha6/biosynthesis , Macrophages/metabolism , Macrophages/pathology , Melanoma, Experimental/pathology , Mice , Mice, Transgenic , Neovascularization, Pathologic/pathology , Receptor, TIE-2/biosynthesis
17.
World Neurosurg ; 82(6): 1171-86, 2014 Dec.
Article in English | MEDLINE | ID: mdl-23178919

ABSTRACT

OBJECTIVE: Monocyte-derived cells of the brain (MDCB) are a diverse group of functional immune cells that are also highly abundant in gliomas. There is growing evidence that MDCB play essential roles in the pathogenesis of gliomas. The aim of this review was to collate and systematize contemporary knowledge about these cells as they relate to glioma progression and antiglioblastoma therapeutic modalities with a view toward improved effectiveness of therapy. METHODS: We reviewed relevant studies to construct a summary of different MDCB subpopulations in steady state and in malignant gliomas and discuss their role in the development of malignant gliomas and potential future therapies. RESULTS: Current studies suggest that MDCB subsets display different phenotypes and differentiation potentials depending on their milieu in the brain and exposure to tumoral influences. MDCB possess specific and unique functions, including those that are protumoral and those that are antitumoral. CONCLUSIONS: Elucidating the role of mononuclear-derived cells associated with gliomas is crucial in designing novel immunotherapy strategies. Much progress is needed to characterize markers to identify cell subsets and their specific regulatory roles. Investigation of MDCB can be clinically relevant. Specific MDCB populations potentially can be used for glioma therapy as a target or as cell vehicles that might deliver cytotoxic substances or processes to the glioma microenvironment.


Subject(s)
Brain Neoplasms/pathology , Brain/cytology , Brain/pathology , Glioma/pathology , Monocytes/pathology , Dendritic Cells/pathology , Humans , Inflammation/pathology , Macrophages/pathology , Microglia/pathology , Myeloid Cells/pathology , Receptor, TIE-2/biosynthesis
18.
Urology ; 82(4): 974.e9-13, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23947989

ABSTRACT

OBJECTIVE: To examine the frequency of tumor-infiltrating Tie-2-expressing monocytes (TEMs) in renal cell carcinoma (RCC) and its association with microvessel density (MVD) and other clinical-pathologic features. MATERIALS AND METHODS: This study enrolled 65 consecutive patients with RCC treated with radical nephrectomy. The frequency of tumor-infiltrating TEMs, which was defined as CD14(+) Tie-2(+) cells, was assessed using flow cytometry. MVD was measured by immunohistochemistry using anti-CD34 antibody. The association between clinicopathologic parameters, MVD, and the frequency of tumor-infiltrating TEMs in RCC was assessed. RESULTS: High frequency of tumor-infiltrating TEMs was significantly associated with advanced stage (P = .018), positive lymph nodes (P = .013), high grade (P = .019), and metastases (P = .006). Correlation analysis revealed that the frequency of TEMs was positively correlated with MVD. CONCLUSION: Our findings revealed a significant association between prognostic tumor features, MVD, and the frequency of tumor-infiltrating TEMs in RCC and indicated that TEMs may play an important role in angiogenesis and progression of RCC.


Subject(s)
Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Monocytes/metabolism , Receptor, TIE-2/biosynthesis , Adult , Aged , Carcinoma, Renal Cell/pathology , Disease Progression , Female , Humans , Kidney Neoplasms/pathology , Male , Middle Aged , Neovascularization, Pathologic
19.
Immunobiology ; 218(11): 1370-5, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23932437

ABSTRACT

Macrophage function is not restricted to the innate and adaptive immune responses, but also includes host defence, wound healing, angiogenesis and homeostatic processes. Within the spectrum of macrophage activation there are two extremes: M1 classically activated macrophages which have a pro-inflammatory phenotype, and M2 alternatively activated macrophages which are pro-angiogenic and anti-inflammatory. An important property of macrophages is their plasticity to switch from one phenotype to the other and they can be defined in their polarisation state at any point between the two extremes. In order to determine what stage of activation macrophages are in, it is essential to profile various phenotypic markers for their identification. This review describes the angiogenic role for myeloid cells: circulating monocytes, Tie-2 expressing monocytes (TEMs), myeloid-derived suppressor cells (MDSCs), tumour associated macrophages (TAMs), and neutrophils. Each cell type is discussed by phenotype, roles within angiogenesis and possible targets as a cell therapy. In addition, we also refer to our own research on myeloid angiogenic cells (MACs), outlining their ability to induce angiogenesis and their similarities to alternatively activated M2 macrophages. MACs significantly contribute to vascular repair through paracrine mechanisms as they lack the capacity to differentiate into endothelial cells. Since MACs also retain plasticity, phenotypic changes can occur according to disease states and the surrounding microenvironment. This pro-angiogenic potential of MACs could be harnessed as a novel cellular therapy for the treatment of ischaemic diseases, such as diabetic retinopathy, hind limb ischaemia and myocardial infarction; however, caution needs to be taken when MACs are delivered into an inflammatory milieu.


Subject(s)
Macrophages/immunology , Monocytes/immunology , Myeloid Cells/immunology , Neovascularization, Pathologic/immunology , Cell Differentiation/immunology , Humans , Immunotherapy/methods , Inflammation/immunology , Macrophage Activation/immunology , Neutrophils/immunology , Receptor, TIE-2/biosynthesis , Receptor, TIE-2/metabolism
20.
Angiogenesis ; 16(4): 921-37, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23881168

ABSTRACT

Notch is an intercellular signaling pathway related mainly to sprouting neo-angiogenesis. The objective of our study was to evaluate the angiogenic mechanisms involved in the vascular augmentation (sprouting/intussusception) after Notch inhibition within perfused vascular beds using the chick area vasculosa and MxCreNotch1(lox/lox) mice. In vivo monitoring combined with morphological investigations demonstrated that inhibition of Notch signaling within perfused vascular beds remarkably induced intussusceptive angiogenesis (IA) with resultant dense immature capillary plexuses. The latter were characterized by 40 % increase in vascular density, pericyte detachment, enhanced vessel permeability, as well as recruitment and extravasation of mononuclear cells into the incipient transluminal pillars (quintessence of IA). Combination of Notch inhibition with injection of bone marrow-derived mononuclear cells dramatically enhanced IA with 80 % increase in vascular density and pillar number augmentation by 420 %. Additionally, there was down-regulation of ephrinB2 mRNA levels consequent to Notch inhibition. Inhibition of ephrinB2 or EphB4 signaling induced some pericyte detachment and resulted in up-regulation of VEGFRs but with neither an angiogenic response nor recruitment of mononuclear cells. Notably, Tie-2 receptor was down-regulated, and the chemotactic factors SDF-1/CXCR4 were up-regulated only due to the Notch inhibition. Disruption of Notch signaling at the fronts of developing vessels generally results in massive sprouting. On the contrary, in the already existing vascular beds, down-regulation of Notch signaling triggered rapid augmentation of the vasculature predominantly by IA. Notch inhibition disturbed vessel stability and led to pericyte detachment followed by extravasation of mononuclear cells. The mononuclear cells contributed to formation of transluminal pillars with sustained IA resulting in a dense vascular plexus without concomitant vascular remodeling and maturation.


Subject(s)
Neovascularization, Pathologic/physiopathology , Receptors, Notch/antagonists & inhibitors , Signal Transduction/drug effects , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Bone Marrow Transplantation , Chemokine CXCL12/biosynthesis , Chemokine CXCL12/genetics , Chick Embryo , Gene Expression Regulation , Leukocytes, Mononuclear/transplantation , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/prevention & control , Oligopeptides/pharmacology , Pericytes/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, EphB2/biosynthesis , Receptor, EphB2/genetics , Receptor, EphB4/biosynthesis , Receptor, EphB4/genetics , Receptor, Notch1/deficiency , Receptor, TIE-2/biosynthesis , Receptor, TIE-2/genetics , Receptors, CXCR4/biosynthesis , Receptors, CXCR4/genetics , Receptors, Notch/physiology , Receptors, Vascular Endothelial Growth Factor/biosynthesis , Receptors, Vascular Endothelial Growth Factor/genetics , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL