101
|
Dai B, Qi J, Liu R, Zhang Y. Eupolyphaga sinensis Walker demonstrates angiogenic activity and inhibits A549 cell growth by targeting the KDR signaling pathway. Mol Med Rep 2014; 10:1590-6. [PMID: 25059654 DOI: 10.3892/mmr.2014.2387] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 12/12/2013] [Indexed: 11/06/2022] Open
Abstract
Eupolyphaga sinensis Walker has been reported to have anticoagulation, antithrombotic, liver protective and antitumor effects. In the present study, the inhibitory effects on proliferation of A549 human non‑small cell lung cancer cells and the underlying mechanisms were examined. Firstly, three solvents, 70% ethanol, distilled water and 95% ethanol, were used to extract Eupolyphaga sinensis Walker. The MTT assay results demonstrated that the 70% ethanol extract more potently reduced the growth of A549 cells and it was therefore adopted in the subsequent experiments. Eupolyphaga sinensis Walker 70% ethanol extract significantly inhibited A549 cell migration in a time‑ and dose‑dependent manner and inhibited human umbilical vein endothelial cell proliferation, migration and tube formation. Furthermore, Eupolyphaga sinensis Walker 70% ethanol extract effectively inhibited blood vessel formation in the established tissue model for angiogenesis. In addition, Eupolyphaga sinensis Walker 70% ethanol extract was demonstrated to inhibit the autophosphorylation of KDR, and downregulate the subsequent activation of AKT and extracellular signal regulated kinase (ERK)1/2 in A549 cells. In conclusion, these findings demonstrated that the antitumor mechanism of Eupolyphaga sinensis Walker 70% ethanol extract was through inhibiting angiogenesis. It functioned by interrupting the autophosphorylation of KDR and subsequently, AKT and ERK1/2.
Collapse
Affiliation(s)
- Bingling Dai
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Junpeng Qi
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Rui Liu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
102
|
Zhang Y, Zhan Y, Zhang D, Dai B, Ma W, Qi J, Liu R, He L. Eupolyphaga sinensis walker displays inhibition on hepatocellular carcinoma through regulating cell growth and metastasis signaling. Sci Rep 2014; 4:5518. [PMID: 24980220 PMCID: PMC4076680 DOI: 10.1038/srep05518] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 06/13/2014] [Indexed: 11/09/2022] Open
Abstract
Tumor growth and metastasis are responsible for most cancer patients' deaths. Here, we report that eupolyphaga sinensis walker has an essential role in resisting hepatocellular carcinoma growth and metastasis. Compared with proliferation, colony formation, transwell assay and transplantable tumor in nude mouse in vitro and vivo, eupolyphaga sinensis walker extract (ESWE) showed good inhibition on the SMMC-7721 cell growth and metastasis. Using genome-wide microarray analysis, we found the down-regulated growth and metastasis factors, and selected down-regulated genes were confirmed by real-time PCR. Knockdown of a checkpoint PKCβ by siRNA significantly attenuated tumor inhibition and metastasis effects of ESWE. Moreover, our results indicate ESWE inhibits HCC growth by not only downregulating the signaling of PKCβ, Akt, m-TOR, Erk1/2, MEK-2, Raf and JNK-1, but also increasing cyclin D1 protein levels and decreasing amount of cyclin E, cyclin B1 and cdc2 of the cycle proteins. At the same time, ESWE reduced MMP2, MMP9 and CXCR4, PLG, NFκB and P53 activities. Overall, our studies demonstrate that ESWE is a key factor in growth and metastasis signaling inhibitor targeting the PKC, AKT, MAPK signaling and related metastasis signaling, having potential in cancer therapy.
Collapse
Affiliation(s)
- Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Yingzhuan Zhan
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Dongdong Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Bingling Dai
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Weina Ma
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Junpeng Qi
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Rui Liu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Langchong He
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| |
Collapse
|
103
|
Marine compound catunaregin inhibits angiogenesis through the modulation of phosphorylation of akt and eNOS in vivo and in vitro. Mar Drugs 2014; 12:2790-801. [PMID: 24824025 PMCID: PMC4052316 DOI: 10.3390/md12052790] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 04/09/2014] [Accepted: 04/14/2014] [Indexed: 11/16/2022] Open
Abstract
Angiogenesis is the formation of blood vessels from pre-existing vasculature. Excessive or uncontrolled angiogenesis is a major contributor to many pathological conditions whereas inhibition of aberrant angiogenesis is beneficial to patients with pathological angiogenesis. Catunaregin is a core of novel marine compound isolated from mangrove associate. The potential anti-angiogenesis of catunaregin was investigated in human umbilical vein endothelial cells (HUVECs) and zebrafish. HUVECs were treated with different concentrations of catunaregin in the presence or absence of VEGF. The angiogenic phenotypes including cell invasion cell migration and tube formation were evaluated following catunaregin treatment in HUVECs. The possible involvement of AKT, eNOS and ERK1/2 in catunaregin-induced anti-angiogenesis was explored using Western blotting. The anti-angiogenesis of catunaregin was further tested in the zebrafish embryo neovascularization and caudal fin regeneration assays. We found that catunaregin dose-dependently inhibited angiogenesis in both HUVECs and zebrafish embryo neovascularization and zebrafish caudal fin regeneration assays. In addition, catunaregin significantly decreased the phosphorylation of Akt and eNOS, but not the phosphorylation of ERK1/2. The present work demonstrates that catunaregin exerts the anti-angiogenic activity at least in part through the regulation of the Akt and eNOS signaling pathways.
Collapse
|
104
|
Abstract
Diabetic retinopathy is a common condition that occurs in patients with diabetes with long-standing hyperglycemia that is characterized by inappropriate angiogenesis. This pathological angiogenesis could be a sort of physiological proliferative response to injury by the endothelium. Recent studies suggested that reactive oxygen species (ROS) play a significant role in this angiogenesis. Vascular endothelial growth factor (VEGF) is a potent angiogenic growth factor that plays a significant role in diabetic retinopathy. The interaction between VEGF and ROS, and theirs in turn with pro- and anti-inflammatory cytokines and anti-inflammatory bioactive lipid molecules such as lipoxins, resolvins, protectins, and maresins is particularly relevant to understand the pathophysiology of diabetic retinopathy and develop future therapeutic interventions.
Collapse
Affiliation(s)
- Qi Ma
- a Department of Food Science and Nutrition , Zhejiang University , Hangzhou , 310029 , China
| | | | | | | |
Collapse
|
105
|
Zuo PY, Chen XL, Lei YH, Liu CY, Liu YW. Growth arrest-specific gene 6 protein promotes the proliferation and migration of endothelial progenitor cells through the PI3K/AKT signaling pathway. Int J Mol Med 2014; 34:299-306. [PMID: 24789534 DOI: 10.3892/ijmm.2014.1754] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 04/07/2014] [Indexed: 11/05/2022] Open
Abstract
Endothelial progenitor cells (EPCs) play an important role in endothelial repair and vascular regeneration. Growth arrest-specific gene 6 (Gas6) is a novel key regulator of the vascular system, which is linked to a number of cardiovascular diseases. However, the effects of Gas6 on EPCs have not been elucidated to date. The present study was designed to determine the biological function of EPCs treated with Gas6 and to eludicate the underlying mechanisms. EPCs were isolated from umbilical cord blood and treated with various concentrations (25, 50, 100 and 200 ng/ml) of Gas6. The proliferation, migration and angiogenesis of the Gas6-treated EPCs were evaluated by MTT assay, Transwell assay and in vitro tube formation assay, respectively. The phosphorylation status of AKT and ERK was evaluated by western blot analysis. The results demonstrated that treatment with Gas6 enhanced the proliferation and migration of the EPCs in a dose-dependent manner. However, Gas6 did not promote the differentiation of EPCs on Matrigel. Gas6 induced the phosphorylation of AKT, but not that of ERK. The enhanced proliferation and migration induced by Gas6 was markedly suppressed by the inhibitor of PI3K but not by that of ERK. These results suggest that Gas6 activates the AKT signaling pathway, which, in turn, promotes the proliferation and migration of EPCs.
Collapse
Affiliation(s)
- Pei-Yuan Zuo
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xing-Lin Chen
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Ying-Hong Lei
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Cheng-Yun Liu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yu-Wei Liu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
106
|
Endothelial PKCα-MAPK/ERK-phospholipase A2 pathway activation as a response of glioma in a triple culture model. A new role for pericytes? Biochimie 2014; 99:77-87. [PMID: 24287292 DOI: 10.1016/j.biochi.2013.11.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 11/13/2013] [Indexed: 01/08/2023]
|
107
|
Mulyasasmita W, Cai L, Hori Y, Heilshorn SC. Avidity-controlled delivery of angiogenic peptides from injectable molecular-recognition hydrogels. Tissue Eng Part A 2014; 20:2102-14. [PMID: 24490588 DOI: 10.1089/ten.tea.2013.0357] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Peptide mimics of growth factors represent an emerging class of therapeutic drugs due to high biological specificity and relative ease of synthesis. However, maintaining efficacious therapeutic dosage at the therapy site has proven challenging owing to poor intestinal permeability and short circulating half-lives in the blood stream. In this work, we present the affinity immobilization and controlled release of QK, a vascular endothelial growth factor (VEGF) mimetic peptide, from an injectable mixing-induced two-component hydrogel (MITCH). The MITCH system is crosslinked by reversible interactions between WW domains and complementary proline-rich peptide modules. Fusion of the QK peptide to either one or two units of the proline-rich sequence creates bifunctional peptide conjugates capable of specific binding to MITCH while preserving their angiogenic bioactivity. Presenting two repeats of the proline-rich sequence increases the binding enthalpy 2.5 times due to avidity effects. Mixing of the drug conjugates with MITCH components results in drug encapsulation and extended release at rates consistent with the affinity immobilization strength. Human umbilical vein endothelial cells (HUVECs) treated with the soluble drug conjugates exhibit morphogenetic events of VEGF receptor 2 signal transduction followed by cell migration and organization into networks characteristic of early angiogenesis. In a three-dimensional model where HUVECs were cultured as spheroids in a matrix of collagen and fibronectin, injection of drug-releasing MITCH resulted in significantly more cell outgrowth than drugs injected in saline. This ability to sustain local drug availability is ideal for therapeutic angiogenesis applications, where spatiotemporal control over drug distribution is a key requirement for clinical success.
Collapse
Affiliation(s)
- Widya Mulyasasmita
- 1 Department of Bioengineering, Stanford University , Stanford, California
| | | | | | | |
Collapse
|
108
|
Pink M, Verma N, Rettenmeier AW, Schmitz-Spanke S. Integrated proteomic and metabolomic analysis to assess the effects of pure and benzo[a]pyrene-loaded carbon black particles on energy metabolism and motility in the human endothelial cell line EA.hy926. Arch Toxicol 2014; 88:913-34. [DOI: 10.1007/s00204-014-1200-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 01/14/2014] [Indexed: 12/20/2022]
|
109
|
The marine fungal metabolite, AD0157, inhibits angiogenesis by targeting the Akt signaling pathway. Mar Drugs 2014; 12:279-99. [PMID: 24441613 PMCID: PMC3917274 DOI: 10.3390/md12010279] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 12/26/2013] [Accepted: 12/27/2013] [Indexed: 11/17/2022] Open
Abstract
In the course of a screening program for the inhibitors of angiogenesis from marine sources, AD0157, a pyrrolidinedione fungal metabolite, was selected for its angiosupressive properties. AD0157 inhibited the growth of endothelial and tumor cells in culture in the micromolar range. Our results show that subtoxic doses of this compound inhibit certain functions of endothelial cells, namely, differentiation, migration and proteolytic capability. Inhibition of the mentioned essential steps of in vitro angiogenesis is in agreement with the observed antiangiogenic activity, substantiated by using two in vivo angiogenesis models, the chorioallantoic membrane and the zebrafish embryo neovascularization assays, and by the ex vivo mouse aortic ring assay. Our data indicate that AD0157 induces apoptosis in endothelial cells through chromatin condensation, DNA fragmentation, increases in the subG1 peak and caspase activation. The data shown here altogether indicate for the first time that AD0157 displays antiangiogenic effects, both in vitro and in vivo, that are exerted partly by targeting the Akt signaling pathway in activated endothelial cells. The fact that these effects are carried out at lower concentrations than those required for other inhibitors of angiogenesis makes AD0157 a new promising drug candidate for further evaluation in the treatment of cancer and other angiogenesis-related pathologies.
Collapse
|
110
|
Liu CL, Kwok HF, Cheng L, Ko CH, Wong CW, Ho TWF, Leung PC, Fung KP, Lau CBS. Molecular mechanisms of angiogenesis effect of active sub-fraction from root of Rehmannia glutinosa by zebrafish sprout angiogenesis-guided fractionation. JOURNAL OF ETHNOPHARMACOLOGY 2014; 151:565-75. [PMID: 24247081 DOI: 10.1016/j.jep.2013.11.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 08/17/2013] [Accepted: 11/10/2013] [Indexed: 05/04/2023]
Abstract
ETHNOPHARMACOLOGICAL IMPORTANCE The root of Rehmannia glutinosa (Rehmanniae Radix (RR)) is clinically used as a wound-healing agent in traditional Chinese medicine. Angiogenesis acts crucially in the pathogenesis of chronic wound healing. The present study investigated the angiogenesis effect and its underlying mechanism of RR through zebrafish sprout angiogenesis guided-fractionation. MATERIALS AND METHODS The in vivo angiogenesis effect was studied by analyzing the number of ectopic sprouts formed upon sub-intestinal vessel of transgenic TG(fli1:EGFP)(y1)/+(AB) zebrafish embryos by fluorescence microscopy. Quantitative real-time PCR gene expression of the zebrafish embryos was further performed using a panel of 30 angiogenesis-associated genes designed for zebrafish sprout angiogenesis. Classical in vitro angiogenesis assays using human microvascular endothelial cells (HMEC-1) was accompanied. RESULTS We demonstrated that among all RR sub-fractions tested, C1-1 treated-zebrafish embryos possessed the most potent angiogenesis activities (from 190 to 780 ng/ml, p<0.001) in sprout formation in the zebrafish model. Quantitative gene expression of the treated embryos demonstrated significant up-regulation in MMP-9 (p<0.05), ANGPT1 (p<0.05), EGFR (p<0.05), EPHB4 (p<0.01), and significant down-regulation in Ephrin B2 (p<0.05), Flt-1 (p<0.05) and Ets-1 (p<0.05). C1-1 treatment could also significantly (p<0.001-0.05) stimulate HMEC-1 cell migration in scratch assay. Significant increase (p<0.05) in mean tubule length was observed in the C1-1-treated HMEC-1 cells in the tubule formation assay. CONCLUSIONS Our zebrafish sprout angiogenesis model-guided fractionation revealed that C1-1 possessed the most potent angiogenesis effect in RR. The design of the panel with 30 tailor-made angiogenesis-associated genes exhibited in zebrafish gene expression analysis showed that C1-1 could trigger differential expression of various angiogenesis-associated genes, such as VEGFR3 and MMP9, which played key role in angiogenesis. The pro-angiogenic activity of C1-1 was further confirmed in the translated study in motogenic and tubule-inducing effect using HMEC-1.
Collapse
Affiliation(s)
- Cheuk-Lun Liu
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong; State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong; School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong
| | - Hin-Fai Kwok
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong; State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong
| | - Ling Cheng
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong; State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong
| | - Chun-Hay Ko
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong; State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong
| | - Chun-Wai Wong
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong; State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong
| | - Tina Wai Fong Ho
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong; State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong
| | - Ping-Chung Leung
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong; State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong
| | - Kwok-Pui Fung
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong; State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong; School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong
| | - Clara Bik-San Lau
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong; State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, Hong Kong.
| |
Collapse
|
111
|
Abdelsaid MA, Matragoon S, El-Remessy AB. Thioredoxin-interacting protein expression is required for VEGF-mediated angiogenic signal in endothelial cells. Antioxid Redox Signal 2013; 19:2199-212. [PMID: 23718729 PMCID: PMC3869450 DOI: 10.1089/ars.2012.4761] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
AIMS Thioredoxin-interacting protein (TXNIP) contributes to cellular redox-state homeostasis via binding and inhibiting thioredoxin (TRX). Increasing evidence suggests that cellular redox homeostasis regulates vascular endothelial growth factor (VEGF)-mediated signaling. This study aims to examine the redox-dependant role of TXNIP in regulating VEGF-mediated S-glutathionylation and angiogenic signaling. TXNIP-knockout mice (TKO) or wild-type (WT) treated with the reduced glutathione (GSH)-precursor, N-acetyl cysteine (WT-NAC, 500 mg/kg) were compared to WT using hypoxia-induced neovascularization model. RESULTS In response to hypoxia, retinas from TKO and WT-NAC mice showed significant decreases in reparative revascularization and pathological neovascularization with similar VEGF expression compared with WT. VEGF failed to stimulate vascular sprouting from aortic rings of TKO compared to WT mice. TKO mice or WT+NAC experienced reductive stress as indicated by twofold increase in TRX reductase activity and fourfold increase in reduced-GSH levels compared with WT. In human microvascular endothelial (HME) cells, VEGF stimulated co-precipitation between vascular endothelial growth factor receptor 2 (VEGFR2) with low molecular weight protein tyrosine phosphatase (LMW-PTP). Silencing TXNIP expression blunted VEGF-induced oxidation of GSH and S-glutathionylation of the LMW-PTP in HME cells. These effects were associated with impaired VEGFR2 phosphorylation that culminated in inhibiting cell migration and tube formation. Overexpression of TXNIP restored VEGFR2 phosphorylation and cell migration in TKO-endothelial cells. INNOVATION TXNIP expression is required for VEGF-mediated VEGFR2 activation and angiogenic response in vivo and in vitro. TXNIP expression regulates VEGFR-2 phosphorylation via S-glutathionylation of LMW-PTP in endothelial cells. CONCLUSION Our results provide novel mechanistic insight into modulating TXNIP expression as a potential therapeutic target in diseases characterized by aberrant angiogenesis.
Collapse
Affiliation(s)
- Mohammed A Abdelsaid
- 1 Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia , Augusta, Georgia
| | | | | |
Collapse
|
112
|
Norisoboldine suppresses VEGF-induced endothelial cell migration via the cAMP-PKA-NF-κB/Notch1 pathway. PLoS One 2013; 8:e81220. [PMID: 24349042 PMCID: PMC3857208 DOI: 10.1371/journal.pone.0081220] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2013] [Accepted: 10/09/2013] [Indexed: 12/11/2022] Open
Abstract
The migration of endothelial cells has been regarded as a potential target for the treatment of angiogenesis-related diseases. Previously, we demonstrated that norisoboldine (NOR), an alkaloid compound isolated from Radix Linderae, can significantly suppress synovial angiogenesis by selectively inhibiting endothelial cell migration. In this study, we evaluated the importance of various pathways in VEGF-induced endothelial cell migration using specific inhibitor. VEGF-induced endothelial cell migration and sprouting were significantly inhibited by H-89 (an inhibitor of protein kinase A (PKA)) but not by inhibitors of other pathways. NOR markedly suppressed VEGF-induced intracytoplasmic cAMP production and PKA activation and thereby down-regulated the activation of downstream components of the PKA pathway, including enzymes (src, VASP and eNOS) and the transcription factor NF-κB. Moreover, the transcription activation potential of NF-κB, which is related to IκBα phosphorylation and the disruption of the p65/IκBα complex, was reduced by NOR. Meanwhile, NOR selectively inhibited the expression of p-p65 (ser276) but not p-p65 (ser536) or PKAc, indicating that PKAc participates in the regulation of NF-κB by NOR. Co-immunoprecipitation and immunofluorescence assays confirmed that NOR inhibited the formation of the PKAc/p65 complex and thereby decreased p65 (ser276) phosphorylation to prevent p65 binding to DNA. Docking models indicated that the affinity of NOR for PKA was higher than that of the original PKA ligand. Moreover, the fact that H-89 improved Notch1 activation, but DAPT (an inhibitor of Notch) failed to affect PKA activation, suggested that PKA may act on upstream of Notch1. In conclusion, the inhibitory effects of NOR on endothelial cell migration can be attributed to its modulation of the PKA pathway, especially on the processes of p65/IκBα complex disruption and PKAc/p65 complex formation. These results suggest that NOR inhibit VEGF-induced endothelial cell migration via a cAMP-PKA-NF-κB/Notch1 signaling pathway.
Collapse
|
113
|
Yu S, Zuo Z, Cui H, Li M, Peng X, Zhu L, Zhang M, Li X, Xu Z, Gan M, Deng J, Fang J, Ma J, Su S, Wang Y, Shen L, Ma X, Ren Z, Wu B, Hu Y. Transcriptional profiling of hilar nodes from pigs after experimental infection with Actinobacillus pleuropneumoniae. Int J Mol Sci 2013; 14:23516-32. [PMID: 24351863 PMCID: PMC3876060 DOI: 10.3390/ijms141223516] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 11/12/2013] [Accepted: 11/15/2013] [Indexed: 11/16/2022] Open
Abstract
The gram-negative bacterium Actinobacillus pleuropneumoniae (APP) is an inhabitant of the porcine upper respiratory tract and the causative agent of porcine pleuropneumonia (PP). In recent years, knowledge about the proinflammatory cytokine and chemokine gene expression that occurs in lung and lymph node of the APP-infected swine has been advanced. However, systematic gene expression profiles on hilar nodes from pigs after infection with Actinobacillus pleuropneumoniae have not yet been reported. The transcriptional responses were studied in hilar nodes (HN) from swine experimentally infected with APP and the control groupusing Agilent Porcine Genechip, including 43,603 probe sets. 9,517 transcripts were identified as differentially expressed (DE) at the p ≤ 0.01 level by comparing the log2 (normalized signal) of the two groups named treatment group (TG) and controls (CG). Eight hundred and fifteen of these DE transcripts were annotated as pig genes in the GenBank database (DB). Two hundred and seventy-two biological process categories (BP), 75 cellular components and 171 molecular functions were substantially altered in the TG compared to CG. Many BP were involved in host immune responses (i.e., signaling, signal transmission, signal transduction, response to stimulus, oxidation reduction, response to stress, immune system process, signaling pathway, immune response, cell surface receptor linked signaling pathway). Seven DE gene pathways (VEGF signaling pathway, Long-term potentiation, Ribosome, Asthma, Allograft rejection, Type I diabetes mellitus and Cardiac muscle contraction) and statistically significant associations with host responses were affected. Many cytokines (including NRAS, PI3K, MAPK14, CaM, HSP27, protein phosphatase 3, catalytic subunit and alpha isoform), mediating the proliferation and migration of endothelial cells and promoting survival and vascular permeability, were activated in TG, whilst many immunomodulatory cytokines were suppressed. The significant changes in the expression patterns of the genes, GO terms, and pathways, led to a decrease of antigenic peptides with antigen presenting cells presented to T lymphocytes via the major histocompatibility complex, and alleviated immune response induced APP of HN. The immune response ability of HN in the APP-infected pigs was weakened; however, cell proliferation and migration ability was enhanced.
Collapse
Affiliation(s)
- Shumin Yu
- College of Veterinary Medicine, Sichuan Agricultural University, Ya’an 625014, China; E-Mails: (S.Y.); (Z.Z.); (X.P.); (L.Z.); (Z.X.); (M.G.); (J.D.); (J.F.); (Y.W.); (L.S.); (X.M.); (Z.R.); (B.W.); (Y.H.)
- Laboratory of Animal Disease and Human Health, Sichuan Agricultural University, Ya’an 625014, China
| | - Zhicai Zuo
- College of Veterinary Medicine, Sichuan Agricultural University, Ya’an 625014, China; E-Mails: (S.Y.); (Z.Z.); (X.P.); (L.Z.); (Z.X.); (M.G.); (J.D.); (J.F.); (Y.W.); (L.S.); (X.M.); (Z.R.); (B.W.); (Y.H.)
- Laboratory of Animal Disease and Human Health, Sichuan Agricultural University, Ya’an 625014, China
| | - Hengmin Cui
- College of Veterinary Medicine, Sichuan Agricultural University, Ya’an 625014, China; E-Mails: (S.Y.); (Z.Z.); (X.P.); (L.Z.); (Z.X.); (M.G.); (J.D.); (J.F.); (Y.W.); (L.S.); (X.M.); (Z.R.); (B.W.); (Y.H.)
- Laboratory of Animal Disease and Human Health, Sichuan Agricultural University, Ya’an 625014, China
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +86-136-0826-4628; Fax: +86-835-2882340
| | - Mingzhou Li
- College of Animal Science and Technology, Sichuan Agricultural University, Ya’an 625014, China; E-Mails: (M.L.); (M.Z.); (X.L.); (J.M.)
| | - Xi Peng
- College of Veterinary Medicine, Sichuan Agricultural University, Ya’an 625014, China; E-Mails: (S.Y.); (Z.Z.); (X.P.); (L.Z.); (Z.X.); (M.G.); (J.D.); (J.F.); (Y.W.); (L.S.); (X.M.); (Z.R.); (B.W.); (Y.H.)
- Laboratory of Animal Disease and Human Health, Sichuan Agricultural University, Ya’an 625014, China
| | - Ling Zhu
- College of Veterinary Medicine, Sichuan Agricultural University, Ya’an 625014, China; E-Mails: (S.Y.); (Z.Z.); (X.P.); (L.Z.); (Z.X.); (M.G.); (J.D.); (J.F.); (Y.W.); (L.S.); (X.M.); (Z.R.); (B.W.); (Y.H.)
- Laboratory of Animal Disease and Human Health, Sichuan Agricultural University, Ya’an 625014, China
| | - Ming Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Ya’an 625014, China; E-Mails: (M.L.); (M.Z.); (X.L.); (J.M.)
| | - Xuewei Li
- College of Animal Science and Technology, Sichuan Agricultural University, Ya’an 625014, China; E-Mails: (M.L.); (M.Z.); (X.L.); (J.M.)
| | - Zhiwen Xu
- College of Veterinary Medicine, Sichuan Agricultural University, Ya’an 625014, China; E-Mails: (S.Y.); (Z.Z.); (X.P.); (L.Z.); (Z.X.); (M.G.); (J.D.); (J.F.); (Y.W.); (L.S.); (X.M.); (Z.R.); (B.W.); (Y.H.)
- Laboratory of Animal Disease and Human Health, Sichuan Agricultural University, Ya’an 625014, China
| | - Meng Gan
- College of Veterinary Medicine, Sichuan Agricultural University, Ya’an 625014, China; E-Mails: (S.Y.); (Z.Z.); (X.P.); (L.Z.); (Z.X.); (M.G.); (J.D.); (J.F.); (Y.W.); (L.S.); (X.M.); (Z.R.); (B.W.); (Y.H.)
- Laboratory of Animal Disease and Human Health, Sichuan Agricultural University, Ya’an 625014, China
| | - Junliang Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Ya’an 625014, China; E-Mails: (S.Y.); (Z.Z.); (X.P.); (L.Z.); (Z.X.); (M.G.); (J.D.); (J.F.); (Y.W.); (L.S.); (X.M.); (Z.R.); (B.W.); (Y.H.)
- Laboratory of Animal Disease and Human Health, Sichuan Agricultural University, Ya’an 625014, China
| | - Jing Fang
- College of Veterinary Medicine, Sichuan Agricultural University, Ya’an 625014, China; E-Mails: (S.Y.); (Z.Z.); (X.P.); (L.Z.); (Z.X.); (M.G.); (J.D.); (J.F.); (Y.W.); (L.S.); (X.M.); (Z.R.); (B.W.); (Y.H.)
- Laboratory of Animal Disease and Human Health, Sichuan Agricultural University, Ya’an 625014, China
| | - Jideng Ma
- College of Animal Science and Technology, Sichuan Agricultural University, Ya’an 625014, China; E-Mails: (M.L.); (M.Z.); (X.L.); (J.M.)
| | - Shengqun Su
- Library of Sichuan Agricultural University, Ya’an 625014, China; E-Mail:
| | - Ya Wang
- College of Veterinary Medicine, Sichuan Agricultural University, Ya’an 625014, China; E-Mails: (S.Y.); (Z.Z.); (X.P.); (L.Z.); (Z.X.); (M.G.); (J.D.); (J.F.); (Y.W.); (L.S.); (X.M.); (Z.R.); (B.W.); (Y.H.)
- Laboratory of Animal Disease and Human Health, Sichuan Agricultural University, Ya’an 625014, China
| | - Liuhong Shen
- College of Veterinary Medicine, Sichuan Agricultural University, Ya’an 625014, China; E-Mails: (S.Y.); (Z.Z.); (X.P.); (L.Z.); (Z.X.); (M.G.); (J.D.); (J.F.); (Y.W.); (L.S.); (X.M.); (Z.R.); (B.W.); (Y.H.)
- Laboratory of Animal Disease and Human Health, Sichuan Agricultural University, Ya’an 625014, China
| | - Xiaoping Ma
- College of Veterinary Medicine, Sichuan Agricultural University, Ya’an 625014, China; E-Mails: (S.Y.); (Z.Z.); (X.P.); (L.Z.); (Z.X.); (M.G.); (J.D.); (J.F.); (Y.W.); (L.S.); (X.M.); (Z.R.); (B.W.); (Y.H.)
- Laboratory of Animal Disease and Human Health, Sichuan Agricultural University, Ya’an 625014, China
| | - Zhihua Ren
- College of Veterinary Medicine, Sichuan Agricultural University, Ya’an 625014, China; E-Mails: (S.Y.); (Z.Z.); (X.P.); (L.Z.); (Z.X.); (M.G.); (J.D.); (J.F.); (Y.W.); (L.S.); (X.M.); (Z.R.); (B.W.); (Y.H.)
- Laboratory of Animal Disease and Human Health, Sichuan Agricultural University, Ya’an 625014, China
| | - Bangyuan Wu
- College of Veterinary Medicine, Sichuan Agricultural University, Ya’an 625014, China; E-Mails: (S.Y.); (Z.Z.); (X.P.); (L.Z.); (Z.X.); (M.G.); (J.D.); (J.F.); (Y.W.); (L.S.); (X.M.); (Z.R.); (B.W.); (Y.H.)
- Laboratory of Animal Disease and Human Health, Sichuan Agricultural University, Ya’an 625014, China
| | - Yanchun Hu
- College of Veterinary Medicine, Sichuan Agricultural University, Ya’an 625014, China; E-Mails: (S.Y.); (Z.Z.); (X.P.); (L.Z.); (Z.X.); (M.G.); (J.D.); (J.F.); (Y.W.); (L.S.); (X.M.); (Z.R.); (B.W.); (Y.H.)
- Laboratory of Animal Disease and Human Health, Sichuan Agricultural University, Ya’an 625014, China
| |
Collapse
|
114
|
Abstract
Increased endothelial permeability and reduction of alveolar liquid clearance capacity are two leading pathogenic mechanisms of pulmonary edema, which is a major complication of acute lung injury, severe pneumonia, and acute respiratory distress syndrome, the pathologies characterized by unacceptably high rates of morbidity and mortality. Besides the success in protective ventilation strategies, no efficient pharmacological approaches exist to treat this devastating condition. Understanding of fundamental mechanisms involved in regulation of endothelial permeability is essential for development of barrier protective therapeutic strategies. Ongoing studies characterized specific barrier protective mechanisms and identified intracellular targets directly involved in regulation of endothelial permeability. Growing evidence suggests that, although each protective agonist triggers a unique pattern of signaling pathways, selected common mechanisms contributing to endothelial barrier protection may be shared by different barrier protective agents. Therefore, understanding of basic barrier protective mechanisms in pulmonary endothelium is essential for selection of optimal treatment of pulmonary edema of different etiology. This article focuses on mechanisms of lung vascular permeability, reviews major intracellular signaling cascades involved in endothelial monolayer barrier preservation and summarizes a current knowledge regarding recently identified compounds which either reduce pulmonary endothelial barrier disruption and hyperpermeability, or reverse preexisting lung vascular barrier compromise induced by pathologic insults.
Collapse
Affiliation(s)
- Konstantin G Birukov
- Lung Injury Center, Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, Illinois, USA.
| | | | | |
Collapse
|
115
|
Lin J, Chen Y, Wei L, Hong Z, Sferra TJ, Peng J. Ursolic acid inhibits colorectal cancer angiogenesis through suppression of multiple signaling pathways. Int J Oncol 2013; 43:1666-1674. [PMID: 24042330 DOI: 10.3892/ijo.2013.2101] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 09/02/2013] [Indexed: 11/05/2022] Open
Abstract
Angiogenesis plays a critical role in the development of solid tumors by supplying nutrients and oxygen to support continuous growth of tumor as well as providing an avenue for hematogenous metastasis. Tumor angiogenesis is highly regulated by multiple intracellular signaling transduction cascades such as Hedgehog, STAT3, Akt and p70S6K pathways that are known to malfunction in many types of cancer including colorectal cancer (CRC). Therefore, suppression of tumor angiogenesis through targeting these signaling pathways has become a promising strategy for cancer chemotherapy. Ursolic acid (UA) is a major active compound present in many medicinal herbs that have long been used in China for the clinical treatment of various types of cancer. Although previous studies have demonstrated an antitumor effect for UA, the precise mechanisms of its anti-angiogenic activity are not well understood. To further elucidate the mechanism(s) of the tumorcidal activity of UA, using a CRC mouse xenograft model, chick embryo chorioallantoic membrane (CAM) model, the human colon carcinoma cell line HT-29 and human umbilical vein endothelial cells (HUVECs), in the present study we evaluated the efficacy of UA against tumor growth and angiogenesis in vivo and in vitro and investigated the underlying molecular mechanisms. We found that administration of UA significantly inhibited tumor volume but had no effect on body weight changes in CRC mice, suggesting that UA can suppress colon cancer growth in vivo without noticeable signs of toxicity. In addition, UA treatment reduced intratumoral microvessel density (MVD) in CRC mice, decreased the total number of blood vessels in the CAM model, and dose and time-dependently inhibited the proliferation, migration and tube formation of HUVECs, demonstrating UA's antitumor angiogenesis in vivo and in vitro. Moreover, UA treatment inhibited the expression of critical angiogenic factors, such as VEGF-A and bFGF. Furthermore, UA suppressed the activation of sonic hedgehog (SHH), STAT3, Akt and p70S6K pathways. Collectively, our findings suggest that inhibition of tumor angiogenesis via suppression of multiple signaling pathways might be one of the mechanisms whereby UA can be effective in cancer treatment.
Collapse
Affiliation(s)
- Jiumao Lin
- Academy of Integrative Medicine Biomedical Research Center, Fujian University of Traditional Chinese Medicine, Minhou Shangjie, Fuzhou, Fujian 350122, P.R. China
| | | | | | | | | | | |
Collapse
|
116
|
Kim HY, Yang DH, Shin SW, Kim MY, Yoon JH, Kim S, Park HC, Kang DW, Min D, Hur MW, Choi KY. CXXC5 is a transcriptional activator of Flk-1 and mediates bone morphogenic protein-induced endothelial cell differentiation and vessel formation. FASEB J 2013; 28:615-26. [PMID: 24136587 DOI: 10.1096/fj.13-236216] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
CXXC5 is a member of a small subset of proteins containing CXXC-type zinc-finger domain. Here, we show that CXXC5 is a transcription factor activating Flk-1, a receptor for vascular endothelial growth factor. CXXC5 and Flk-1 were accumulated in nucli and membrane of mouse embryonic stem cells (mESCs), respectively, during their endothelial differentiation. CXXC5 overexpression induced Flk-1 transcription in both endothelium-differentiated mESCs and human umbilical vein endothelial cells (HUVECs). In vitro DNA binding assay showed direct interaction of CXXC5 on the Flk-1 promoter region, and mutation on its DNA-binding motif abolished transcriptional activity. We showed that bone morphorgenetic protein 4 (BMP4) induced CXXC5 transcription in the cells, and inhibitors of BMP signaling suppressed the CXXC5 induction and the consequent Flk-1 induction by BMP4 treatment. CXXC5 knockdown resulted in suppression of BMP4-induced stress fiber formation (56.8 ± 1.3% decrease, P<0.05) and migration (54.6 ± 1.9% decrease, P<0.05) in HUVECs. The in vivo roles of CXXC5 in BMP-signaling-specific vascular development and angiogenesis were shown by specific defect of caudal vein plex vessel formation (57.9 ± 11.8% decrease, P<0.05) in cxxc5 morpholino-injected zebrafish embryos and by suppression of BMP4-induced angiogenesis in subcutaneously injected Matrigel plugs in CXXC5(-/-) mice. Overall, CXXC5 is a transcriptional activator for Flk-1, mediating BMP signaling for differentiation and migration of endothelial cell and vessel formation.
Collapse
Affiliation(s)
- Hyun-Yi Kim
- 2Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 120-749, South Korea.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
117
|
Luo W, Wang X, Zheng L, Zhan Y, Zhang D, Zhang J, Zhang Y. Brucine suppresses colon cancer cells growth via mediating KDR signalling pathway. J Cell Mol Med 2013; 17:1316-24. [PMID: 23905676 PMCID: PMC4159018 DOI: 10.1111/jcmm.12108] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 07/01/2013] [Indexed: 01/30/2023] Open
Abstract
Angiogenesis plays an important role in colon cancer development. This study aimed to demonstrate the effect of brucine on tumour angiogenesis and its mechanism of action. The anti-angiogenic effect was evaluated on the chicken chorioallantoic membrane (CAM) model and tube formation. The mechanism was demonstrated through detecting mRNA and protein expressions of VEGFR2 (KDR), PKCα, PLCγ and Raf1 by reverse transcription-polymerase chain reaction (RT-PCR) and Western blot (WB), as well as expressions of VEGF and PKCβ and mTOR by ELISA and WB. The results showed that brucine significantly reduced angiogenesis of CAM and tube formation, inhibited the VEGF secretion and mTOR expression in LoVo cell and down-regulated the mRNA and phosphorylation protein expressions of KDR, PKCα, PLCγ and Raf1. In addition, the effects of brucine on KDR kinase activity, viability of LoVo cell and gene knockdown cell were detected with the Lance™ assay, WST-1 assay and instantaneous siRNA. Compared to that of normal LoVo cells, the inhibition on proliferation of knockdown cells by brucine decreased significantly. These results suggest that brucine could inhibit angiogenesis and be a useful therapeutic candidate for colon cancer intervention.
Collapse
Affiliation(s)
- Wenjuan Luo
- School of Medicine, Xi'an Jiaotong UniversityXi'an, Shaanxi Province, China
| | - Xiaoli Wang
- School of Medicine, Xi'an Jiaotong UniversityXi'an, Shaanxi Province, China
| | - Lei Zheng
- School of Medicine, Xi'an Jiaotong UniversityXi'an, Shaanxi Province, China
| | - Yingzhuan Zhan
- School of Medicine, Xi'an Jiaotong UniversityXi'an, Shaanxi Province, China
| | - Dongdong Zhang
- School of Medicine, Xi'an Jiaotong UniversityXi'an, Shaanxi Province, China
| | - Jie Zhang
- School of Medicine, Xi'an Jiaotong UniversityXi'an, Shaanxi Province, China
| | - Yanmin Zhang
- School of Medicine, Xi'an Jiaotong UniversityXi'an, Shaanxi Province, China
| |
Collapse
|
118
|
Bosutti A, Qi J, Pennucci R, Bolton D, Matou S, Ali K, Tsai LH, Krupinski J, Petcu EB, Montaner J, Al Baradie R, Caccuri F, Caruso A, Alessandri G, Kumar S, Rodriguez C, Martinez-Gonzalez J, Slevin M. Targeting p35/Cdk5 signalling via CIP-peptide promotes angiogenesis in hypoxia. PLoS One 2013; 8:e75538. [PMID: 24098701 PMCID: PMC3787057 DOI: 10.1371/journal.pone.0075538] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 08/19/2013] [Indexed: 01/19/2023] Open
Abstract
Cyclin-dependent kinase-5 (Cdk5) is over-expressed in both neurons and microvessels in hypoxic regions of stroke tissue and has a significant pathological role following hyper-phosphorylation leading to calpain-induced cell death. Here, we have identified a critical role of Cdk5 in cytoskeleton/focal dynamics, wherein its activator, p35, redistributes along actin microfilaments of spreading cells co-localising with p(Tyr15)Cdk5, talin/integrin beta-1 at the lamellipodia in polarising cells. Cdk5 inhibition (roscovitine) resulted in actin-cytoskeleton disorganisation, prevention of protein co-localization and inhibition of movement. Cells expressing Cdk5 (D144N) kinase mutant, were unable to spread, migrate and form tube-like structures or sprouts, while Cdk5 wild-type over-expression showed enhanced motility and angiogenesis in vitro, which was maintained during hypoxia. Gene microarray studies demonstrated myocyte enhancer factor (MEF2C) as a substrate for Cdk5-mediated angiogenesis in vitro. MEF2C showed nuclear co-immunoprecipitation with Cdk5 and almost complete inhibition of differentiation and sprout formation following siRNA knock-down. In hypoxia, insertion of Cdk5/p25-inhibitory peptide (CIP) vector preserved and enhanced in vitro angiogenesis. These results demonstrate the existence of critical and complementary signalling pathways through Cdk5 and p35, and through which coordination is a required factor for successful angiogenesis in sustained hypoxic condition.
Collapse
Affiliation(s)
- Alessandra Bosutti
- School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom
| | - Jie Qi
- School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom
| | - Roberta Pennucci
- Cell Adhesion Unit, Department of Neuroscience Dibit-Istituto Scientifico San Raffaele, Milano, Italy
| | | | - Sabine Matou
- School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom
| | - Kamela Ali
- School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom
| | - Li-Huei Tsai
- Howard Hughes Medical Institute, Massachusetts Institute of Technology Picower Institute for Learning and Memory, Cambridge, Massachusetts, United States of America
- Stanley Centre for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, United States of America
| | - Jerzy Krupinski
- School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom
- Hospital Universitari Mútua de Terrassa, Department of Neurology, Barcelona, Spain
| | - Eugene B. Petcu
- Griffith University School of Medicine, Gold Coast Campus, Griffith University, Southport, Australia
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall De’Hebron University Hospital, Barcelona, Spain
| | - Raid Al Baradie
- College of Applied Medical Science, Almajmaah University, Almajmaah, Kingdom of Saudi Arabia
| | - Francesca Caccuri
- University of Brescia, Section of Microbiology, Department of Experimental and Applied Medicine, Medical School, Brescia, Italy
| | - Arnaldo Caruso
- University of Brescia, Section of Microbiology, Department of Experimental and Applied Medicine, Medical School, Brescia, Italy
| | - Giulio Alessandri
- Fondazione Istituto di Ricovero e Cura Carattere Scientifico Neurological Institute "Carlo Besta", Cellular Neurobiology Laboratory, Department of Cerebrovascular Diseases, Milan, Italy
| | - Shant Kumar
- School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom
- Department of Pathological Sciences, Manchester University and Christie Hospital, Manchester, United Kingdom
| | - Cristina Rodriguez
- Centro de Investigacion Cardiovascular, Hospital de la Santa Creu i Sant, Pau, Barcelona, Spain
| | - Jose Martinez-Gonzalez
- Centro de Investigacion Cardiovascular, Hospital de la Santa Creu i Sant, Pau, Barcelona, Spain
| | - Mark Slevin
- School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom
- Griffith University School of Medicine, Gold Coast Campus, Griffith University, Southport, Australia
- *E-mail:
| |
Collapse
|
119
|
Eguchi R, Kubo S, Ohta T, Kunimasa K, Okada M, Tamaki H, Kaji K, Wakabayashi I, Fujimori Y, Ogawa H. FK506 induces endothelial dysfunction through attenuation of Akt and ERK1/2 independently of calcineurin inhibition and the caspase pathway. Cell Signal 2013; 25:1731-8. [DOI: 10.1016/j.cellsig.2013.05.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 05/06/2013] [Indexed: 01/30/2023]
|
120
|
Lu Q, Wang C, Pan R, Gao X, Wei Z, Xia Y, Dai Y. Histamine synergistically promotes bFGF-induced angiogenesis by enhancing VEGF production via H1 receptor. J Cell Biochem 2013; 114:1009-19. [PMID: 23225320 DOI: 10.1002/jcb.24440] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Accepted: 10/24/2012] [Indexed: 11/06/2022]
Abstract
Histamine, a major mediator present in mast cells that is released into the extracellular milieu upon degranulation, is well known to possess a wide range of biological activities in several classic physiological and pathological processes. However, whether and how it participates in angiogenesis remains obscure. In the present study, we observed its direct and synergistic action with basic fibroblast growth factor (bFGF), an important inducer of angiogenesis, on in vitro angiogenesis models of endothelial cells. Data showed that histamine (0.1, 1, 10 µM) itself was absent of direct effects on the processes of angiogenesis, including the proliferation, migration, and tube formation of endothelial cells. Nevertheless, it could concentration-dependently enhance bFGF-induced angiogenesis as well as production of vascular endothelial growth factor (VEGF) from endothelial cells. The synergistic effect of histamine on VEGF production could be reversed by pretreatments with diphenhydramine (H1-receptor antagonist), SB203580 (selective p38 mitogen-activated protein kinase (MAPK) inhibitor) and L-NAME (nitric oxide synthase (NOS) inhibitor), but not with cimetidine (H2-receptor antagonist) and indomethacin (cyclooxygenase (COX) inhibitor). Moreover, histamine could augment bFGF-incuced phosphorylation and degradation of IκBα, a key factor accounting for the activation and translocation of nuclear factor κB (NF-κB) in endothelial cells. These findings indicated that histamine was able to synergistically augment bFGF-induced angiogenesis, and this action was linked to VEGF production through H1-receptor and the activation of endothelial nitric oxide synthase (eNOS), p38 MAPK, and IκBα in endothelial cells.
Collapse
Affiliation(s)
- Qian Lu
- Department of Pharmacology of Chinese Materia Medica, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | | | | | | | | | | | | |
Collapse
|
121
|
Kang Z, Jiang W, Luan H, Zhao F, Zhang S. Cornin induces angiogenesis through PI3K–Akt–eNOS–VEGF signaling pathway. Food Chem Toxicol 2013; 58:340-6. [DOI: 10.1016/j.fct.2013.05.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Revised: 05/09/2013] [Accepted: 05/10/2013] [Indexed: 01/05/2023]
|
122
|
Rennier K, Ji JY. The role of death-associated protein kinase (DAPK) in endothelial apoptosis under fluid shear stress. Life Sci 2013; 93:194-200. [DOI: 10.1016/j.lfs.2013.06.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 05/30/2013] [Accepted: 06/13/2013] [Indexed: 01/13/2023]
|
123
|
Krstić J, Jauković A, Mojsilović S, Ðorđević IO, Trivanović D, Ilić V, Santibañez JF, Bugarski D. In vitro effects of IL-17 on angiogenic properties of endothelial cells in relation to oxygen levels. Cell Biol Int 2013; 37:1162-70. [PMID: 23765637 DOI: 10.1002/cbin.10144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 06/05/2013] [Indexed: 12/20/2022]
Abstract
The aim of this study has been to elucidate how different oxygen levels impact the effects of Interleukin-17 (IL-17) on angiogenic properties of endothelial cells. Two endothelial cell lines, mouse MS-1 and human EA.hy 926, were grown in 20% and 3% O2 and their angiogenic abilities analyzed after IL-17 treatment: proliferation, apoptosis, migration and tubulogenesis. Expression of endothelial nitric oxide synthase (eNOS) and cyclooxygenase-2 (Cox-2) was also measured. Considering EA.hy 926 cell line, hypoxia alone reduced proliferation, survival and migration, but not their ability to form tubules. When cultured at 20% O2 , IL-17 stimulated proliferation, migration and tubulogenesis, whereas a hypoxic environment did not affect their migration and proliferation, but increased their survival and tubulogenic properties. Expression of eNOS and Cox-2 increased by both IL-17 and hypoxia, as well as with their combination. With the MS-1 cell line hypoxia did not affect proliferation, survival, migration and tubule formation. At 20% O2 , IL-17 did not alter their proliferation,but inhibited migration and stimulated tubule formation. At 3% O2 , only the stimulating effect of IL-17 on tubulogenesis was evident. The constitutive expression of eNOS was unaffected by oxygen concentrations or IL-17 supplementation, whereas both IL-17 and hypoxia upregulated Cox-2 expression. Thus the effects of IL-17 on the angiogenic properties of endothelial cells depend on both the cell line used and the oxygen concentration.
Collapse
Affiliation(s)
- Jelena Krstić
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Dr. Subotića 4, P.O. Box 102, 11129, Belgrade, Serbia
| | | | | | | | | | | | | | | |
Collapse
|
124
|
Kang Z, Zhu H, Jiang W, Zhang S. Protocatechuic acid induces angiogenesis through PI3K-Akt-eNOS-VEGF signalling pathway. Basic Clin Pharmacol Toxicol 2013; 113:221-7. [PMID: 23738793 DOI: 10.1111/bcpt.12094] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 05/06/2013] [Indexed: 10/26/2022]
Abstract
In this study, we sought to elucidate whether protocatechuic acid contributes to induce angiogenesis as well as its mechanisms. To this end, we examined the role of protocatechuic acid on human brain microvascular endothelial cell line (HBMEC) proliferation, invasion and tube formation in in vitro. For the study of mechanisms involved, the phosphoinositide 3 kinase (PI3K)-Akt inhibitor LY294002, the endothelial nitric oxide synthase (eNOS) inhibitor L-NAME, vascular endothelial growth factor (VEGF), antagonist sFlt-1 and VEGF receptor blocker SU-1498 were used. Proliferation of HBMEC was tested by MTT. Scratch adhesion test was used to assess the ability of invasion. A Matrigel tube formation assay was performed to test capillary tube formation ability. PI3K-Akt-eNOS-VEGF pathway activation in HBMEC was tested by Western blot. Our data suggested that protocatechuic acid induces angiogenesis in vitro by increasing proliferation, invasion and tube formation. VEGF expression was increasing by protocatechuic acid and counteracted by VEGF antagonist sFlt-1, LY294002 and L-NAME in HBMEC. Tube formation was increased by protocatechuic acid and counteracted by VEGF receptor blocker-SU1498, LY294002 and L-NAME. These data suggest that protocatechuic acid may be a candidate therapy for stroke recovery by promoting angiogenesis via a programmed PI3K/Akt/eNOS/VEGF signalling axis.
Collapse
Affiliation(s)
- Zechun Kang
- School of Pharmaceutical Sciences and Institute of Material Medica, Binzhou Medical University, Yantai, China
| | | | | | | |
Collapse
|
125
|
Liu CL, Tam JCW, Sanders AJ, Ko CH, Fung KP, Leung PC, Harding KG, Jiang WG, Lau CBS. Molecular angiogenic events of a two-herb wound healing formula involving MAPK and Akt signaling pathways in human vascular endothelial cells. Wound Repair Regen 2013; 21:579-87. [DOI: 10.1111/wrr.12055] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 03/25/2013] [Indexed: 11/26/2022]
Affiliation(s)
| | | | - Andrew J. Sanders
- Metastasis and Angiogenesis Research Group; Cardiff University School of Medicine; Cardiff; United Kingdom
| | | | | | | | - Keith G. Harding
- Department of Dermatology and Wound Healing; Cardiff University School of Medicine; Cardiff; United Kingdom
| | - Wen G. Jiang
- Metastasis and Angiogenesis Research Group; Cardiff University School of Medicine; Cardiff; United Kingdom
| | | |
Collapse
|
126
|
Yang P, Zhang Y, Pang J, Zhang S, Yu Q, He L, Wagner KU, Zhou Z, Wang CY. Loss of Jak2 impairs endothelial function by attenuating Raf-1/MEK1/Sp-1 signaling along with altered eNOS activities. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:617-25. [PMID: 23747947 DOI: 10.1016/j.ajpath.2013.04.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 02/17/2013] [Accepted: 04/08/2013] [Indexed: 10/26/2022]
Abstract
A number of inhibitors have been used to dissect the functional relevance of Jak2 in endothelial homeostasis, with disparate results. Given that Jak2 deficiency leads to embryonic lethality, the exact role of Jak2 in the regulation of postnatal endothelial function is yet to be fully elucidated. We generated a model in which Jak2 deficiency can be induced by tamoxifen in adult mice. Loss of Jak2 significantly impaired endothelium-dependent response capacity for vasodilators. Matrigel plug assays indicated a notable decrease in endothelial angiogenic function in Jak2-deficient mice. Studies in a hindlimb ischemic model indicated that Jak2 activity is likely to be a prerequisite for prompt perfusion recovery, based on the concordance of temporal changes in Jak2 expression during the course of ischemic injury and perfusion recovery. A remarkable delay in perfusion recovery, along with reduced capillary and arteriole formation, was observed in Jak2-deficient mice. Antibody array studies indicated that loss of Jak2 led to repressed eNOS expression. In mechanistic studies, Jak2 deficiency attenuated Raf-1/MEK1 signaling, which then reduced activity of Sp-1, an essential transcription factor responsible for eNOS expression. These data are important not only for understanding the exact role that Jak2 plays in endothelial homeostasis, but also for assessing Jak2-based therapeutic strategies in a variety of clinical settings.
Collapse
Affiliation(s)
- Ping Yang
- Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | | | | | |
Collapse
|
127
|
Butein Inhibits Angiogenesis of Human Endothelial Progenitor Cells via the Translation Dependent Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:943187. [PMID: 23840271 PMCID: PMC3690248 DOI: 10.1155/2013/943187] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 05/09/2013] [Indexed: 12/17/2022]
Abstract
Compelling evidence indicates that bone marrow-derived endothelial progenitor cells (EPCs) can contribute to postnatal neovascularization and tumor angiogenesis. EPCs have been shown to play a "catalytic" role in metastatic progression by mediating the angiogenic switch. Understanding the pharmacological functions and molecular targets of natural products is critical for drug development. Butein, a natural chalcone derivative, has been reported to exert potent anticancer activity. However, the antiangiogenic activity of butein has not been addressed. In this study, we found that butein inhibited serum- and vascular endothelial growth factor- (VEGF-) induced cell proliferation, migration, and tube formation of human EPCs in a concentration dependent manner without cytotoxic effect. Furthermore, butein markedly abrogated VEGF-induced vessels sprouting from aortic rings and suppressed microvessel formation in the Matrigel implant assay in vivo. In addition, butein concentration-dependently repressed the phosphorylation of Akt, mTOR, and the major downstream effectors, p70S6K, 4E-BP1, and eIF4E in EPCs. Taken together, our results demonstrate for the first time that butein exhibits the antiangiogenic effect both in vitro and in vivo by targeting the translational machinery. Butein is a promising angiogenesis inhibitor with the potential for treatment of cancer and other angiogenesis-related diseases.
Collapse
|
128
|
Li H, Chang J. Bioactive silicate materials stimulate angiogenesis in fibroblast and endothelial cell co-culture system through paracrine effect. Acta Biomater 2013; 9:6981-91. [PMID: 23416471 DOI: 10.1016/j.actbio.2013.02.014] [Citation(s) in RCA: 196] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 01/09/2013] [Accepted: 02/07/2013] [Indexed: 01/23/2023]
Abstract
Angiogenesis is critical in tissue engineering, and bioceramic-induced angiogenesis has been reported. However, the role of other types of cells such as fibroblasts in this bioceramic-induced angiogenesis process has not been reported, and is closer to the in vivo situation of tissue regeneration. In this study, the paracrine effect of silicate bioceramic-induced angiogenesis in the presence of fibroblasts was confirmed by investigating the effects of calcium silicate (CS), one of the simplest silicate bioactive ceramics, on angiogenesis in co-cultures of human dermal fibroblasts (HDF) and human umbilical vein endothelial cells (HUVEC). Results showed that CS extracts stimulated the expression of vascular endothelial growth factor (VEGF) from co-cultured HDF and subsequently enhanced the expression of VEGF receptor 2 on co-cultured HUVEC (co-HUVEC). The endothelial nitric oxide synthase and nitric oxide production in co-HUVEC was then increased to finally initiate the proangiogenesis. During this process, the expression of vascular endothelial cadherin from co-HUVEC was up-regulated, and cadherin proteins were concentrated at the cell junctions to facilitate tube formation. Silicon ions are confirmed to play an important role during silicate bioceramic-inducing angiogenesis, and effective silicon ion concentrations (0.7-1.8μgml(-1)) are proposed.
Collapse
Affiliation(s)
- H Li
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai 200030, China
| | | |
Collapse
|
129
|
Serum Containing Tao-Hong-Si-Wu Decoction Induces Human Endothelial Cell VEGF Production via PI3K/Akt-eNOS Signaling. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:195158. [PMID: 23762109 PMCID: PMC3673321 DOI: 10.1155/2013/195158] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Revised: 04/11/2013] [Accepted: 04/21/2013] [Indexed: 01/05/2023]
Abstract
Tao-Hong-Si-Wu decoction (TSD) is a famous traditional Chinese medicine (TCM) and widely used for ischemic disease in China. TSD medicated serum was prepared after oral administration of TSD (1.6 g/kg) twice a day for 3 days in rats. TSD medicated serum induced human umbilical vein endothelial cells (HUVECs) proliferation, VEGF secretion, and nitric oxide (NO) production. These promoted effects of TSD were partly inhibited by treatment with PI3K inhibitor (LY294002) or eNOS inhibitor (L-NAME), respectively, and completely inhibited by treatment with LY294002 and L-NAME simultaneously. Western blot analysis findings further indicated that TSD medicated serum upregulated p-Akt and p-eNOS expressions, which were significantly inhibited by LY294002 or L-NAME and completely inhibited by both LY294002 and L-NAME; these results indicated that TSD medicated serum induced HUVECs VEGF expression via PI3K/Akt-eNOS signaling. TSD medicated serum contains hydroxysafflor yellow A, ferulic acid, and ligustilide detected by UPLC with standards, so these effect of TSD medicated serum may be associated with these three active compounds absorbed in serum.
Collapse
|
130
|
Heinke J, Juschkat M, Charlet A, Mnich L, Helbing T, Bode C, Patterson C, Moser M. Antagonism and synergy between extracellular BMP modulators Tsg and BMPER balance blood vessel formation. J Cell Sci 2013; 126:3082-94. [PMID: 23641068 DOI: 10.1242/jcs.122333] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Growth and regeneration of blood vessels are crucial processes during embryonic development and in adult disease. Members of the bone morphogenetic protein (BMP) family are growth factors known to play a key role in vascular development. The BMP pathway is controlled by extracellular BMP modulators such as BMP endothelial cell precursor derived regulator (BMPER), which we reported previously acts proangiogenically on endothelial cells in a concentration-dependent manner. Here, we explore the function of other BMP modulators, especially Tsg, on endothelial cell behaviour and compare them to BMPER. In Matrigel assays, BMP modulators chordin and noggin had no stimulatory effect; however, gremlin and Tsg enhanced human umbilical vein endothelial cell (HUVEC) sprouting. As the activation dynamics of Tsg were similar to those of BMPER, we further investigated the proangiogenic effect of Tsg on endothelial cells. Tsg enhanced endothelial cell ingrowth in the mouse Matrigel plug assay as well as HUVEC sprouting, migration and proliferation in vitro, dependent on Akt, Erk and Smad signalling pathway activation in a concentration-dependent manner. Surprisingly, silencing of Tsg also increased HUVEC sprouting, migration and proliferation, which is again associated with Akt, Erk and Smad signalling pathway activation. Furthermore, we reveal that Tsg and BMPER interfere with each other to enhance proangiogenic events. However, in vivo the presence of Tsg as well as of BMPER is mandatory for regular development of the zebrafish vasculature. Taken together, our results suggest that BMPER and Tsg maintain a fine-tuned equilibrium that controls BMP pathway activity and is necessary for vascular cell homeostasis.
Collapse
Affiliation(s)
- Jennifer Heinke
- Heart Center, Freiburg University, Cardiology and Angiology I, 79106 Freiburg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
131
|
Wen HC, Kao C, Hsu RC, Huo YN, Ting PC, Chen LC, Hsu SP, Juan SH, Lee WS. Thy-1-induced migration inhibition in vascular endothelial cells through reducing the RhoA activity. PLoS One 2013; 8:e61506. [PMID: 23613866 PMCID: PMC3629179 DOI: 10.1371/journal.pone.0061506] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 03/10/2013] [Indexed: 11/18/2022] Open
Abstract
Our previous study indicated that Thy-1, which is expressed on blood vessel endothelium in settings of pathological and a specific of physiological, but not during embryonic, angiogenesis, may be used as a marker for angiogenesis. However, the function of Thy-1 during angiogenesis is still not clear. Here, we demonstrate that knock-down of the endogenous Thy-1 expression by Thy-1 siRNA transfection promoted the migration of human umbilical vein endothelial cells (HUVEC). In contrast, treatment with interleukin-1β (IL-1β) or phorbol-12-myristate-13-acetate (PMA) increased the level of Thy-1 protein and reduced the migration of HUVEC. These effects were abolished by pre-transfection of HUVEC with Thy-1 siRNA to knock-down the expression of Thy-1. Moreover, over-expression of Thy-1 by transfection of HUVEC with Thy-1 pcDNA3.1 decreased the activity of RhoA and Rac-1 and inhibited the adhesion, migration and capillary-like tube formation of these cells. These effects were prevented by co-transfection of the cell with constitutively active RhoA construct (RhoA V14). On the other hand, pre-treatment with a ROCK (a kinase associated with RhoA for transducing RhoA signaling) inhibitor, Y27632, abolished the RhoA V14-induced prevention effect on the Thy-1-induced inhibition of endothelial cell migration and tube formation. Taken together, these results indicate that suppression of the RhoA-mediated pathway might participate in the Thy-1-induced migration inhibition in HUVEC. In the present study, we uncover a completely novel role of Thy-1 in endothelial cell behaviors.
Collapse
Affiliation(s)
- Heng-Ching Wen
- Graduate Institute of Medical Sciences, Medical College, Taipei Medical University, Taipei, Taiwan
| | - Chieh Kao
- Graduate Institute of Cell and Molecular Biology, Medical College, Taipei Medical University, Taipei, Taiwan
| | - Ruei-Chi Hsu
- Graduate Institute of Medical Sciences, Medical College, Taipei Medical University, Taipei, Taiwan
| | - Yen-Nien Huo
- Graduate Institute of Medical Sciences, Medical College, Taipei Medical University, Taipei, Taiwan
| | - Pei-Ching Ting
- Graduate Institute of Medical Sciences, Medical College, Taipei Medical University, Taipei, Taiwan
| | - Li-Ching Chen
- Graduate Institute of Medical Sciences, Medical College, Taipei Medical University, Taipei, Taiwan
| | - Sung-Po Hsu
- Department of Physiology, School of Medicine, Medical College, Taipei Medical University, Taipei, Taiwan
| | - Shu-Hui Juan
- Department of Physiology, School of Medicine, Medical College, Taipei Medical University, Taipei, Taiwan
| | - Wen-Sen Lee
- Graduate Institute of Medical Sciences, Medical College, Taipei Medical University, Taipei, Taiwan
- Department of Physiology, School of Medicine, Medical College, Taipei Medical University, Taipei, Taiwan
- Cancer Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| |
Collapse
|
132
|
García-Caballero M, Marí-Beffa M, Cañedo L, Medina MÁ, Quesada AR. Toluquinol, a marine fungus metabolite, is a new angiosuppresor that interferes with the Akt pathway. Biochem Pharmacol 2013; 85:1727-40. [PMID: 23603293 DOI: 10.1016/j.bcp.2013.04.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 04/08/2013] [Accepted: 04/09/2013] [Indexed: 10/26/2022]
Abstract
Toluquinol, a methylhydroquinone produced by a marine fungus, was selected in the course of a blind screening for new potential inhibitors of angiogenesis. In the present study we provide the first evidence that toluquinol is a new anti-angiogenic-compound. In a variety of experimental systems, representing the sequential events of the angiogenic process, toluquinol treatment of activated endothelial cells resulted in strong inhibitory effect. Toluquinol inhibited the growth of endothelial and tumor cells in culture in the micromolar range. Our results indicate that the observed growth inhibitory effect could be due, at least in part, to an induction of apoptosis. Toluquinol induced endothelial cell death is mediated via apoptosis after a cell cycle block and caspase activation. Capillary tube formation on Matrigel and migratory, invasive and proteolytic capabilities of endothelial cells were inhibited by addition of toluquinol at subtoxic concentrations. Inhibition of the mentioned essential steps of in vitro angiogenesis agrees with the observed inhibition of the in vivo angiogenesis, substantiated by using the chick chorioallatoic membrane assay and confirmed by the murine Matrigel plug, the zebrafish embryo neovascularization and the zebrafish caudal fin regeneration assays. Data here shown altogether indicate that toluquinol has antiangiogenic effects both in vitro and in vivo that are exerted partly by suppression of the VEGF and FGF-induced Akt activation of endothelial cells. These effects are carried out at lower concentrations to those required for other inhibitors of angiogenesis, what makes toluquinol a promising drug candidate for further evaluation in the treatment of cancer and other angiogenesis-related pathologies.
Collapse
Affiliation(s)
- Melissa García-Caballero
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Málaga, E-29071 Málaga, Spain
| | | | | | | | | |
Collapse
|
133
|
Scott JM, Lakoski S, Mackey JR, Douglas PS, Haykowsky MJ, Jones LW. The potential role of aerobic exercise to modulate cardiotoxicity of molecularly targeted cancer therapeutics. Oncologist 2013; 18:221-31. [PMID: 23335619 PMCID: PMC3579607 DOI: 10.1634/theoncologist.2012-0226] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Accepted: 09/05/2012] [Indexed: 01/03/2023] Open
Abstract
Molecularly targeted therapeutics (MTT) are the future of cancer systemic therapy. They have already moved from palliative therapy for advanced solid malignancies into the setting of curative-intent treatment for early-stage disease. Cardiotoxicity is a frequent and potentially serious adverse complication of some targeted therapies, leading to a broad range of potentially life-threatening complications, therapy discontinuation, and poor quality of life. Low-cost pleiotropic interventions are therefore urgently required to effectively prevent and/or treat MTT-induced cardiotoxicity. Aerobic exercise therapy has the unique capacity to modulate, without toxicity, multiple gene expression pathways in several organ systems, including a plethora of cardiac-specific molecular and cell-signaling pathways implicated in MTT-induced cardiac toxicity. In this review, we examine the molecular signaling of antiangiogenic and HER2-directed therapies that may underpin cardiac toxicity and the hypothesized molecular mechanisms underlying the cardioprotective properties of aerobic exercise. It is hoped that this knowledge can be used to maximize the benefits of small molecule inhibitors, while minimizing cardiac damage in patients with solid malignancies.
Collapse
Affiliation(s)
- Jessica M Scott
- Exercise Physiology and Countermeasures, NASA Johnson Space Center, Universities Space Research Association, 2101 NASA Parkway, Houston, TX 77058, USA.
| | | | | | | | | | | |
Collapse
|
134
|
Zheng X, Young Koh G, Jackson T. A continuous model of angiogenesis: Initiation, extension, and maturation of new blood vessels modulated by vascular endothelial growth factor, angiopoietins, platelet-derived growth factor-B, and pericytes. ACTA ACUST UNITED AC 2013. [DOI: 10.3934/dcdsb.2013.18.1109] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
135
|
Chen ML, Lin YH, Yang CM, Hu ML. Lycopene inhibits angiogenesis both in vitro and in vivo by inhibiting MMP-2/uPA system through VEGFR2-mediated PI3K-Akt and ERK/p38 signaling pathways. Mol Nutr Food Res 2012; 56:889-99. [PMID: 22707264 DOI: 10.1002/mnfr.201100683] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
SCOPE Limited in vitro data show that lycopene may be anti-angiogenic but with unclear mechanisms. Here, we employed ex vivo and in vivo assays to substantiate the anti-angiogenic action of lycopene and determined its molecular mechanisms in human umbilical vein endothelial cells (HUVECs). METHODS AND RESULTS The anti-angiogenic activity of lycopene was confirmed by ex vivo rat aortic ring and in vivo chorioallantoic membrane assays. Furthermore, the in vivo matrigel plug assay in mice demonstrated that lycopene implanted s.c. at the highest dose used (400 μg/plug) completely inhibited the formation of vascular endothelial cells induced by vascular endothelial growth factor (VEGF). As expected, lycopene inhibited tube formation, invasion, and migration in HUVECs, and such actions were accompanied by reduced activities of matrix metalloproteinase-2, urokinase-type plasminogen activator, and protein expression of Rac1, and by enhancing protein expression of tissue inhibitors of metalloproteinase-2 and plasminogen activator inhibitor-1. Moreover, lycopene attenuated VEGF receptor-2 (VEGFR2)-mediated phosphorylation of extracellular signal-regulated kinase (ERK), p38, and Akt as well as protein expression of PI3K. CONCLUSION Our data demonstrate the anti-angiogenic effect of lycopene both in vitro and in vivo. The anti-angiogenic activity of lycopene may involve inhibition of MMP-2/uPA system through VEGFR2-mediated PI3K-Akt and ERK/p38 signaling pathways.
Collapse
Affiliation(s)
- Man-Ling Chen
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | | | | | | |
Collapse
|
136
|
Zhang YM, Dai BL, Zheng L, Zhan YZ, Zhang J, Smith WW, Wang XL, Chen YN, He LC. A novel angiogenesis inhibitor impairs lovo cell survival via targeting against human VEGFR and its signaling pathway of phosphorylation. Cell Death Dis 2012; 3:e406. [PMID: 23059825 PMCID: PMC3481133 DOI: 10.1038/cddis.2012.145] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Colorectal cancer represents the fourth commonest malignancy, and constitutes a major cause of significant morbidity and mortality among other diseases. However, the chemical therapy is still under development. Angiogenesis plays an important role in colon cancer development. We developed HMQ18–22 (a novel analog of taspine) with the aim to target angiogenesis. We found that HMQ18–22 significantly reduced angiogenesis of chicken chorioallantoic membrane (CAM) and mouse colon tissue, and inhibited cell migration and tube formation as well. Then, we verified the interaction between HMQ18–22 and VEGFR2 by AlphaScreen P-VEGFR assay, screened the targets on angiogenesis by VEGF Phospho Antibody Array, validated the target by western blot and RNAi in lovo cells. We found HMQ18–22 could decrease phosphorylation of VEGFR2(Tyr1214), VEGFR1(Tyr1333), Akt(Tyr326), protein kinase Cα (PKCα) (Tyr657) and phospholipase-Cγ-1 (PLCγ-1) (Tyr771). Most importantly, HMQ18–22 inhibited proliferation of lovo cell and tumor growth in a human colon tumor xenografted model of athymic mice. Compared with normal lovo cells proliferation, the inhibition on proliferation of knockdown cells (VEGFR2, VEGFR1, Akt, PKCα and PLCγ-1) by HMQ18–22 decreased. These results suggested that HMQ18–22 is a novel angiogenesis inhibitor and can be a useful therapeutic candidate for colon cancer intervention.
Collapse
Affiliation(s)
- Y M Zhang
- Institute of Materia Medica, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | | | | | | | | | | | | | | | | |
Collapse
|
137
|
Zhang N, Gong L, Zhang H, Cao C. High Glucose–Induced Dysfunction of Endothelial Cells can be Restored by HoxA9EC. Ann Vasc Surg 2012; 26:1002-10. [DOI: 10.1016/j.avsg.2012.05.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 05/22/2012] [Accepted: 05/24/2012] [Indexed: 11/26/2022]
|
138
|
Yi EY, Kim YJ. Betaine inhibits in vitro and in vivo angiogenesis through suppression of the NF-κB and Akt signaling pathways. Int J Oncol 2012; 41:1879-85. [PMID: 22940742 DOI: 10.3892/ijo.2012.1616] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 07/23/2012] [Indexed: 11/06/2022] Open
Abstract
Angiogenesis is defined as the formation of new blood vessels form existing vessels surrounding a tumor. The process of angiogenesis is an important step for tumor growth and metastasis, as is inflammation. Thus, angiogenesis inhibitors that suppress inflammation have been studied as an anticancer treatment. Recently, many research groups have investigated the anti-angiogenic activity of natural compounds since some have been demonstrated to have anticancer properties. Among many natural compounds, we focused on betaine, which is known to suppress inflammation. Betaine, trimethylglycine (TMG), was first discovered in the juice of sugar beets and was later shown to be present in wheat, shellfish and spinach. In Southeast Asia, betaine is used in traditional oriental medicine for the treatment of hepatic disorders. Here, we report the anti-angiogenic action of betaine. Betaine inhibited in vitro angiogenic cascade, tube formation, migration and invasion of human umbilical vein endothelial cells (HUVECs). Betaine also inhibited in vivo angiogenesis in the mouse Matrigel plug assay. The mRNA expression levels of basic fibroblast growth factor (bFGF), matrix metalloproteinase-2 (MMP-2) and matrix metalloproteinase-9 (MMP-9) in HUVECs were decreased by betaine treatment. In addition, betaine suppressed NF-κB and Akt activation.
Collapse
Affiliation(s)
- Eui-Yeun Yi
- Department of Molecular Biology, Pusan National University, Busan 609-735, Republic of Korea
| | | |
Collapse
|
139
|
Kim KJ, Choi JS, Kim KW, Jeong JW. The anti-angiogenic activities of glycyrrhizic acid in tumor progression. Phytother Res 2012; 27:841-6. [PMID: 22899320 DOI: 10.1002/ptr.4800] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 07/03/2012] [Accepted: 07/16/2012] [Indexed: 12/24/2022]
Abstract
Glycyrrhizic acid (GA) is the bioactive compound of licorice and has been used as a herbal medicine because of its anti-viral, anti-cancer, and anti-inflammatory properties. This study was designed to investigate the effects of GA on tumor growth, angiogenesis, and the mechanisms underlying the anti-angiogenic activities of GA. We observed that GA inhibited tumor growth and angiogenesis in mice. GA decreased angiogenic activities, such as the migration, invasion, and tube formation of endothelial cells. We also demonstrated that GA reduced the production of reactive oxygen species and activation of ERK in endothelial cells. Our findings suggest that GA is a promising anti-angiogenic therapeutic agent that targets the ERK pathway. Considering that angiogenesis is highly stimulated in the majority of cancers, GA could offer a potent therapeutic agent for cancer.
Collapse
Affiliation(s)
- Kil-Jung Kim
- Department of Biomedical Science, Kyung Hee University, Seoul, Korea
| | | | | | | |
Collapse
|
140
|
Lu Q, Lu S, Gao X, Luo Y, Tong B, Wei Z, Lu T, Xia Y, Chou G, Wang Z, Dai Y. Norisoboldine, an alkaloid compound isolated from Radix Linderae, inhibits synovial angiogenesis in adjuvant-induced arthritis rats by moderating Notch1 pathway-related endothelial tip cell phenotype. Exp Biol Med (Maywood) 2012; 237:919-32. [PMID: 22875342 DOI: 10.1258/ebm.2012.011416] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Synovial angiogenesis is well recognized as participating in the pathogenesis of rheumatoid arthritis (RA) and has been regarded as a potential target for RA therapy. Previously, we have shown that norisoboldine (NOR) can protect joints from destruction in mice with collagen II-induced arthritis (CIA). Here, we investigate the effect of NOR on synovial angiogenesis in adjuvant-induced arthritis (AA) rats, and clarify the mechanisms in vitro. NOR, administered orally, significantly reduced the number of blood vessels and expression of growth factors in the synovium of AA rats. In vitro, it markedly prevented the migration and sprouting of endothelial cells. Notably, the endothelial tip cell phenotype, which is essential for the migration of endothelial cells and subsequent angiogenesis, was significantly inhibited by NOR. This inhibitory effect was attenuated by pretreatment with N-{N-[2-(3,5-difluorophenyl) acetyl]-(S)-alanyl}-(S)-phenylglycine tert-butyl ester, a Notch1 inhibitor, suggesting that the action of NOR was related to the Notch1 pathway. A molecular docking study further confirmed that NOR was able to promote Notch1 activation by binding the Notch1 transcription complex. In conclusion, NOR was able to prevent synovial angiogenesis in AA rats, which is a putatively new mechanism responsible for its anti-rheumatoid effect. The anti-angiogenesis action of NOR was likely achieved by moderating the Notch1 pathway-related endothelial tip cell phenotype with a potential action target of the Notch1 transcription complex.
Collapse
Affiliation(s)
- Qian Lu
- Department of Pharmacology of Chinese Materia Medica, State Key Laboratory of Natural Medicines
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Zamani A, Qu Z. Serotonin activates angiogenic phosphorylation signaling in human endothelial cells. FEBS Lett 2012; 586:2360-5. [DOI: 10.1016/j.febslet.2012.05.047] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 05/22/2012] [Indexed: 10/28/2022]
|
142
|
Shen AL, Hong F, Liu LY, Lin JM, Zhuang QC, Hong ZF, Peng J. Effects of Pien Tze Huang on angiogenesis in vivo and in vitro. Chin J Integr Med 2012; 18:431-436. [PMID: 22821655 DOI: 10.1007/s11655-012-1121-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To investigate the anti-angiogenic effects of Pien Tze Huang in vivo and in vitro. METHODS Human umbilical vein endothelial cells (HUVECs) were treated with 0 mg/mL, 0.25 mg/mL, 0.5 mg/mL, and 1 mg/mL of PZH for 24 h, 48 h and 72 h, respectively. Chicken embryo chorioallantoic membrane (CAM) model was used to evaluate in vivo angiogenesis. An ECMatrix gel system was used to evaluate in vitro angiogenesis by examining the tube formation of HUVECs. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay was performed to determine HUVEC viability. Cell density of HUVECs was observed by phase-contrast microscopy. HUVEC migration was determined by wound healing method. The mRNA and protein expression of vascular endothelial growth factor A (VEGF-A) and basic fibroblast growth factor (bFGF) in both HUVEC and human colon adenocarcinoma cells (HT-29) was examined by reverse transcription polymerase chain reaction (RT-PCR) and enzyme linked immune sorbent assay (ELISA), respectively. RESULTS PZH treatment significantly reduced the total number of blood vessels compared with the untreated control in the chicken embryos and resulted in a significant decrease in capillary tube formation and cell density of HUVECs (P<0.05). In addition, treatment with 0.25-1 mg/mL of PZH for 24 h, 48 h, and 72 h respectively reduced cell viability by 9%-52%, 24%-87% or 25%-87%, compared with the untreated control cells (P<0.05). Moreover, PZH treatment decreased the migration of HUVECs. Furthermore, PZH dose-dependently suppressed the expression of VEGF-A and bFGF on both mRNA and protein levels (P<0.05). CONCLUSION PZH could inhibit angiogenesis in vivo in CAM model and in vitro on HUVECs, suggesting that inhibiting tumor angiogenesis might be one of the mechanisms by which PZH treats cancer.
Collapse
Affiliation(s)
- A-ling Shen
- Academy of Integrative Medicine Biomedical Research Center, Fujian University of Traditional Chinese Medicine, Fuzhou (350108), China
| | | | | | | | | | | | | |
Collapse
|
143
|
Lai SL, Cheah SC, Wong PF, Noor SM, Mustafa MR. In vitro and in vivo anti-angiogenic activities of Panduratin A. PLoS One 2012; 7:e38103. [PMID: 22666456 PMCID: PMC3364190 DOI: 10.1371/journal.pone.0038103] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Accepted: 05/03/2012] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Targeting angiogenesis has emerged as an attractive and promising strategy in anti-cancer therapeutic development. The present study investigates the anti-angiogenic potential of Panduratin A (PA), a natural chalcone isolated from Boesenbergia rotunda by using both in vitro and in vivo assays. METHODOLOGY/PRINCIPAL FINDINGS PA exerted selective cytotoxicity on human umbilical vein endothelial cells (HUVECs) with IC(50) value of 6.91 ± 0.85 µM when compared to human normal fibroblast and normal liver epithelial cells. Assessment of the growth kinetics by cell impedance-based Real-Time Cell Analyzer showed that PA induced both cytotoxic and cytostatic effects on HUVECs, depending on the concentration used. Results also showed that PA suppressed VEGF-induced survival and proliferation of HUVECs. Furthermore, endothelial cell migration, invasion, and morphogenesis or tube formation demonstrated significant time- and dose-dependent inhibition by PA. PA also suppressed matrix metalloproteinase-2 (MMP-2) secretion and attenuated its activation to intermediate and active MMP-2. In addition, PA suppressed F-actin stress fiber formation to prevent migration of the endothelial cells. More importantly, anti-angiogenic potential of PA was also evidenced in two in vivo models. PA inhibited neo-vessels formation in murine Matrigel plugs, and angiogenesis in zebrafish embryos. CONCLUSIONS/SIGNIFICANCE Taken together, our study demonstrated the distinctive anti-angiogenic properties of PA, both in vitro and in vivo. This report thus reveals another biological activity of PA in addition to its reported anti-inflammatory and anti-cancer activities, suggestive of PA's potential for development as an anti-angiogenic agent for cancer therapy.
Collapse
Affiliation(s)
- Siew-Li Lai
- Centre of Natural Products & Drug Discovery (CENAR), Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Shiau-Chuen Cheah
- Centre of Natural Products & Drug Discovery (CENAR), Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Pooi-Fong Wong
- Centre of Natural Products & Drug Discovery (CENAR), Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Suzita Mohd Noor
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Mohd Rais Mustafa
- Centre of Natural Products & Drug Discovery (CENAR), Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
144
|
Simão F, Pagnussat AS, Seo JH, Navaratna D, Leung W, Lok J, Guo S, Waeber C, Salbego CG, Lo EH. Pro-angiogenic effects of resveratrol in brain endothelial cells: nitric oxide-mediated regulation of vascular endothelial growth factor and metalloproteinases. J Cereb Blood Flow Metab 2012; 32:884-95. [PMID: 22314268 PMCID: PMC3345913 DOI: 10.1038/jcbfm.2012.2] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Resveratrol may be a powerful way of protecting the brain against a wide variety of stress and injury. Recently, it has been proposed that resveratrol not only reduces brain injury but also promotes recovery after stroke. But the underlying mechanisms are unclear. Here, we tested the hypothesis that resveratrol promotes angiogenesis in cerebral endothelial cells and dissected the signaling pathways involved. Treatment of cerebral endothelial cells with resveratrol promoted proliferation, migration, and tube formation in Matrigel assays. Consistent with these pro-angiogenic responses, resveratrol altered endothelial morphology resulting in cytoskeletal rearrangements of β-catenin and VE-cadherin. These effects of resveratrol were accompanied by activation of phosphoinositide 3 kinase (PI3-K)/Akt and Mitogen-Activated Protein Kinase (MAPK)/ERK signaling pathways that led to endothelial nitric oxide synthase upregulation and increased nitric oxide (NO) levels. Subsequently, elevated NO signaling increased vascular endothelial growth factor and matrix metalloproteinase levels. Sequential blockade of these signaling steps prevented resveratrol-induced angiogenesis in cerebral endothelial cells. These findings provide a mechanistic basis for the potential use of resveratrol as a candidate therapy to promote angiogenesis and neurovascular recovery after stroke.
Collapse
Affiliation(s)
- Fabricio Simão
- Departments of Radiology and Neurology, Neuroprotection Research Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
145
|
Criswell KA, Cook JC, Morse D, Lawton M, Somps C, Obert L, Roy M, Sokolowski S, Koza-Taylor P, Colangelo J, Navetta K, Brady J, Pegg D, Wojcinski Z, Rahbari R, Duddy S, Anderson T. Pregabalin Induces Hepatic Hypoxia and Increases EndothelialCell Proliferation in Mice, a Process Inhibited by DietaryVitamin E Supplementation. Toxicol Sci 2012; 128:42-56. [DOI: 10.1093/toxsci/kfs148] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
146
|
Neuropilin-1 is upregulated in Sjögren's syndrome and contributes to pathological neovascularization. Histochem Cell Biol 2012; 137:669-77. [PMID: 22237885 DOI: 10.1007/s00418-012-0910-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/31/2011] [Indexed: 12/21/2022]
Abstract
Neuropilin-1 (NRP1) is a transmembrane co-receptor for members of the vascular endothelial growth factor family. Recent studies revealed an important role of NRP1 in angiogenesis and progression of many diseases. The role of NRP1 in the development of Sjögren's syndrome (SS), one of the most common rheumatic diseases, has not yet been investigated. Molecular studies and protein expression techniques were performed to elucidate the gene and protein expression profile of NRP1 in human salivary gland epithelial cells (SGEC) from primary SS. We used human microarrays and transient transfection with a mutant form of the negative inhibitory κBα proteins (IκBαDN) to investigate whether selective inhibition of nuclear Factor-κB (NF-κB) improves NRP1-mediated pro-angiogenic factors release from SS SGEC. The selective NRP1 function inhibition with an antibody to human NRP1, was employed to evaluate the therapeutic potential of targeting NRP1. We demonstrate that NRP1 is expressed in SGEC of both human healthy biopsies and in SS samples, and increased NRP1 expression in SS SGEC is significantly associated with pro-angiogenic factors release. Neutralizing anti-NRP1 antibody decreased pro-angiogenic factor production from SS SGEC and blocking NF-κB activation could be a way to inhibit NRP1-mediated angiogenesis in Sjögren's syndrome.
Collapse
|
147
|
|
148
|
Hedhli N, Dobrucki LW, Kalinowski A, Zhuang ZW, Wu X, Russell RR, Sinusas AJ, Russell KS. Endothelial-derived neuregulin is an important mediator of ischaemia-induced angiogenesis and arteriogenesis. Cardiovasc Res 2011; 93:516-24. [PMID: 22200588 DOI: 10.1093/cvr/cvr352] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
AIMS Neuregulins (NRG) are growth factors that are synthesized by endothelial cells (ECs) and bind to erbB receptors. We have shown previously that NRG is proangiogenic in vitro, and that NRG/erbB signalling is important for autocrine endothelial angiogenic signalling in vitro. However, the role of NRG in the angiogenic response to ischaemia is unknown. We hypothesized that endothelial NRG is required for ischaemia-induced angiogenesis in vivo and that exogenous administration of NRG will enhance angiogenic responses after ischaemic insult. METHODS AND RESULTS An endothelial-selective inducible NRG knockout mouse was created and subjected to femoral artery ligation. Endothelial NRG deletion significantly decreased blood flow recovery (by 40%, P < 0.05), capillary density, α(v)β(3) integrin activation, and arteriogenesis after ischaemic injury. Isolated ECs from knockout mice demonstrated significantly impaired cord formation in vitro, suggesting that NRG signalling performs an important cell autonomous function. Recombinant human NRG (rNRG) has not only reversed the angiogenic defect in knockout mice but also accelerated blood flow recovery in wild-type mice. CONCLUSION Endothelial production of NRG is required for angiogenesis and arteriogenesis induced by ischaemic injury. Furthermore, exogenous administration of rNRG can enhance this process, suggesting a potential role for NRG in vascular disease.
Collapse
Affiliation(s)
- Nadia Hedhli
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT, USA
| | | | | | | | | | | | | | | |
Collapse
|
149
|
López-Jiménez A, García-Caballero M, Medina MÁ, Quesada AR. Anti-angiogenic properties of carnosol and carnosic acid, two major dietary compounds from rosemary. Eur J Nutr 2011; 52:85-95. [DOI: 10.1007/s00394-011-0289-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Accepted: 11/28/2011] [Indexed: 12/14/2022]
|
150
|
Kuo MW, Wang CH, Wu HC, Chang SJ, Chuang YJ. Soluble THSD7A is an N-glycoprotein that promotes endothelial cell migration and tube formation in angiogenesis. PLoS One 2011; 6:e29000. [PMID: 22194972 PMCID: PMC3237571 DOI: 10.1371/journal.pone.0029000] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 11/18/2011] [Indexed: 12/19/2022] Open
Abstract
Background Thrombospondin type I domain containing 7A (THSD7A) is a novel neural protein that is known to affect endothelial migration and vascular patterning during development. To further understand the role of THSD7A in angiogenesis, we investigated the post-translational modification scheme of THS7DA and to reveal the underlying mechanisms by which this protein regulates blood vessel growth. Methodology/Principal Findings Full-length THSD7A was overexpressed in human embryonic kidney 293T (HEK293T) cells and was found to be membrane associated and N-glycosylated. The soluble form of THSD7A, which is released into the cultured medium, was harvested for further angiogenic assays. We found that soluble THSD7A promotes human umbilical vein endothelial cell (HUVEC) migration and tube formation. HUVEC sprouts and zebrafish subintestinal vessel (SIV) angiogenic assays further revealed that soluble THSD7A increases the number of branching points of new vessels. Interestingly, we found that soluble THSD7A increased the formation of filopodia in HUVEC. The distribution patterns of vinculin and phosphorylated focal adhesion kinase (FAK) were also affected, which implies a role for THSD7A in focal adhesion assembly. Moreover, soluble THSD7A increased FAK phosphorylation in HUVEC, suggesting that THSD7A is involved in regulating cytoskeleton reorganization. Conclusions/Significance Taken together, our results indicate that THSD7A is a membrane-associated N-glycoprotein with a soluble form. Soluble THSD7A promotes endothelial cell migration during angiogenesis via a FAK-dependent mechanism and thus may be a novel neuroangiogenic factor.
Collapse
Affiliation(s)
- Meng-Wei Kuo
- Department of Medical Science and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
| | - Chian-Huei Wang
- Department of Medical Science and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
| | - Hsiao-Chun Wu
- Department of Medical Science and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
| | - Shing-Jyh Chang
- Department of Obstetrics and Gynecology, Hsinchu Mackay Memorial Hospital, Hsinchu, Taiwan, Republic of China
| | - Yung-Jen Chuang
- Department of Medical Science and Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
- * E-mail:
| |
Collapse
|