201
|
Jung B, Yang EJ, Bae JS. Suppressive effects of polyozellin on TGFBIp-mediated septic responses in human endothelial cells and mice. Nutr Res 2015; 36:380-389. [PMID: 27001283 DOI: 10.1016/j.nutres.2015.12.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 12/08/2015] [Accepted: 12/16/2015] [Indexed: 10/22/2022]
Abstract
Polyozellus multiplex (Thelephoraceae) is a wild mushroom in Korea and Japan and is usually harvested in early autumn for food. Polyozellin, a major constituent of the edible mushroom P multiplex, has been known to exhibit biological activities such as antioxidative and anti-inflammatory effects. Transforming growth factor β-induced protein (TGFBIp) is an extracellular matrix protein whose expression in several cell types is greatly increased by TGF-β. TGFBIp is released by human umbilical vein endothelial cells and functions as a mediator of experimental sepsis. We hypothesized that polyozellin could reduce TGFBIp-mediated severe inflammatory responses in human endothelial cells and mice. Here, we investigated the antiseptic effects and underlying mechanisms of polyozellin against TGFBIp-mediated septic responses. Polyozellin effectively inhibited lipopolysaccharide-induced release of TGFBIp and suppressed TGFBIp-mediated septic responses. In addition, polyozellin suppressed cecal ligation and puncture-induced sepsis lethality and pulmonary injury. In conclusion, polyozellin suppressed TGFBIp-mediated and cecal ligation and puncture-induced septic responses. Therefore, polyozellin could be a potential therapeutic agent for treatment of various severe vascular inflammatory diseases via inhibition of the TGFBIp signaling pathway.
Collapse
Affiliation(s)
- Byeongjin Jung
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Eun-Ju Yang
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Jong-Sup Bae
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea.
| |
Collapse
|
202
|
Bertin FR, Lemarié CA, Robins RS, Blostein MD. Growth arrest-specific 6 regulates thrombin-induced expression of vascular cell adhesion molecule-1 through forkhead box O1 in endothelial cells. J Thromb Haemost 2015; 13:2260-72. [PMID: 26414399 DOI: 10.1111/jth.13156] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 09/12/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND Growth arrest-specific 6 (Gas6)-deficient mice are protected against venous thromboembolism (VTE), suggesting a role for Gas6 in this disorder. We previously demonstrated that Gas6 induces forkhead box O1 (FoxO-1) phosphorylation through the phosphoinositide 3-kinase-Akt pathway. FoxO-1 regulates the expression of vascular cell adhesion molecule-1 (VCAM-1), a molecule that has been implicated in VTE. OBJECTIVES To assess the role of FoxO-1 in Gas6-dependent VCAM-1 expression. METHODS Thrombin was used to stimulate endothelial cells (ECs). Wild-type (WT) and Gas6(-/-) ECs were transfected with small interfering RNA targeting Axl or FoxO-1, a luciferase-coupled plasmid containing the FoxO-1 consensus sequence, and a phosphorylation-resistant FoxO-1 mutant, or treated with an Akt inhibitor. VCAM-1 mRNA expression was measured by real time-qPCR. VCAM-1 protein expression and FoxO-1 and Akt phosphorylation were assessed by western blot analysis. FoxO-1 localization was assessed by immunofluorescence. Adhesion of bone marrow mononuclear cells (BM-MCs) on ECs was assessed by fluorescence. RESULTS AND CONCLUSIONS Thrombin induces both VCAM-1 expression and FoxO-1 phosphorylation and nuclear exclusion in WT ECs only. Silencing of FoxO-1 enhances VCAM-1 expression in both WT and Gas6(-/-) ECs. Inhibition of Akt or FoxO-1 phosphorylation prevents VCAM-1 expression in WT ECs. These data show that Gas6 induces FoxO-1 phosphorylation, leading to derepression of VCAM-1 expression. BM-MC-EC adhesion is increased by thrombin in WT ECs. BM-MC-EC adhesion is further increased when FoxO-1 is silenced, but decreased when FoxO-1 phosphorylation is inhibited. These results demonstrate that the Gas6-FoxO-1 signaling axis plays an important role in VCAM-1 expression in the context of VTE by promoting BM-MC-EC adhesion.
Collapse
Affiliation(s)
- F R Bertin
- Lady Davis Institute for Medical Research, McGill University, Montreal, Quebec, Canada
| | - C A Lemarié
- Lady Davis Institute for Medical Research, McGill University, Montreal, Quebec, Canada
- Department of Medicine, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - R S Robins
- The Royal Victoria Hospital, McGill University Health Center, McGill University, Montreal, Quebec, Canada
| | - M D Blostein
- Lady Davis Institute for Medical Research, McGill University, Montreal, Quebec, Canada
- Department of Medicine, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
203
|
Sambri I, Crespo J, Aguiló S, Ingrosso D, Rodríguez C, Martínez González J. miR-17 and -20a Target the Neuron-Derived Orphan Receptor-1 (NOR-1) in Vascular Endothelial Cells. PLoS One 2015; 10:e0141932. [PMID: 26600038 PMCID: PMC4658114 DOI: 10.1371/journal.pone.0141932] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/14/2015] [Indexed: 11/19/2022] Open
Abstract
Neuron-derived orphan receptor-1 (NOR-1) plays a major role in vascular biology by controlling fibroproliferative and inflammatory responses. Because microRNAs (miRNAs) have recently emerged as key players in the regulation of gene expression in the vasculature, here we have investigated the regulation of NOR-1 by miRNAs in endothelial cells. Computational algorithms suggest that NOR-1 could be targeted by members of the miR-17 family. Accordingly, ectopic over-expression of miR-17 or miR-20a in endothelial cells using synthetic premiRNAs attenuated the up-regulation of NOR-1 expression induced by VEGF (as evidenced by real time PCR, Western blot and immunocitochemistry). Conversely, the antagonism of these miRNAs by specific antagomirs prevented the down-regulation of NOR-1 promoted by miR-17 or miR-20a in VEGF-stimulated cells. Disruption of the miRNA-NOR-1 mRNA interaction using a custom designed target protector evidenced the selectivity of these responses. Further, luciferase reporter assays and seed-sequence mutagenesis confirmed that miR-17 and -20a bind to NOR-1 3’-UTR. Finally, miR-17 and -20a ameliorated the up-regulation of VCAM-1 mediated by NOR-1 in VEGF-stimulated cells. Therefore, miR-17 and -20a target NOR-1 thereby regulating NOR-1-dependent gene expression.
Collapse
Affiliation(s)
- Irene Sambri
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
- Department of Biochemistry, Biophysics & General Pathology, School of Medicine & Surgery, Second University of Naples, Naples, Italy
| | - Javier Crespo
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - Silvia Aguiló
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - Diego Ingrosso
- Department of Biochemistry, Biophysics & General Pathology, School of Medicine & Surgery, Second University of Naples, Naples, Italy
| | - Cristina Rodríguez
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - José Martínez González
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
- * E-mail:
| |
Collapse
|
204
|
The Neuropilin-1 Inhibitor, ATWLPPR Peptide, Prevents Experimental Diabetes-Induced Retinal Injury by Preserving Vascular Integrity and Decreasing Oxidative Stress. PLoS One 2015; 10:e0142571. [PMID: 26554379 PMCID: PMC4640834 DOI: 10.1371/journal.pone.0142571] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 10/24/2015] [Indexed: 12/26/2022] Open
Abstract
Neuropilin-1 (NRP-1) is a transmembrane glycoprotein. As a VEGF co-receptor, NRP1 significantly enhances VEGFR2 signaling and promotes vascular permeability and migration. The purpose of this study was to evaluate the effects of an NRP-1 inhibitor, ATWLPPR peptide, on the early stages of diabetic retinopathy. Eight-week-old male C57BL/6 mice were divided into three groups: a Normal group, a Diabetes (DB) ATWLPPR treatment group and a DB saline group. Electroretinography (ERG), fundus fluorescence angiography (FFA) and leukostasis were examined to evaluate the retinal injury induced by diabetes at the end of the fifth week after STZ injection. Occludin expression and extravasation of albumin were measured to determine the extent of vascular injury. The oxidative stress level and the levels of inflammation-associated proteins were also assayed. The results indicated that treatment with ATWLPPR prevents the abnormal condition of ERG (amplitudes of b-wave decreased and implicit time increased) and vascular injury (occludin degradation and increase in extravasated albumin). These effects were associated with a reduction in the oxidase stress level and the expression of VEGF, GFAP, and ICAM-1. We conclude that ATWLPPR, an NRP-1 inhibitor, may reduce the early retinal damage induced by diabetes by preserving vascular integrity and decreasing the oxidative stress level. Blockade of NRP-1 may be a new therapeutic strategy for the early stages of DR.
Collapse
|
205
|
Khan MA, Hsu JL, Assiri AM, Broering DC. Targeted complement inhibition and microvasculature in transplants: a therapeutic perspective. Clin Exp Immunol 2015; 183:175-86. [PMID: 26404106 DOI: 10.1111/cei.12713] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2015] [Indexed: 12/18/2022] Open
Abstract
Active complement mediators play a key role in graft-versus-host diseases, but little attention has been given to the angiogenic balance and complement modulation during allograft acceptance. The complement cascade releases the powerful proinflammatory mediators C3a and C5a anaphylatoxins, C3b, C5b opsonins and terminal membrane attack complex into tissues, which are deleterious if unchecked. Blocking complement mediators has been considered to be a promising approach in the modern drug discovery plan, and a significant number of therapeutic alternatives have been developed to dampen complement activation and protect host cells. Numerous immune cells, especially macrophages, develop both anaphylatoxin and opsonin receptors on their cell surface and their binding affects the macrophage phenotype and their angiogenic properties. This review discusses the mechanism that complement contributes to angiogenic injury, and the development of future therapeutic targets by antagonizing activated complement mediators to preserve microvasculature in rejecting the transplanted organ.
Collapse
Affiliation(s)
- M A Khan
- Organ Transplant Centre, Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| | - J L Hsu
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - A M Assiri
- Organ Transplant Centre, Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| | - D C Broering
- Organ Transplant Centre, Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
206
|
Chen GF, Sudhahar V, Youn SW, Das A, Cho J, Kamiya T, Urao N, McKinney RD, Surenkhuu B, Hamakubo T, Iwanari H, Li S, Christman JW, Shantikumar S, Angelini GD, Emanueli C, Ushio-Fukai M, Fukai T. Copper Transport Protein Antioxidant-1 Promotes Inflammatory Neovascularization via Chaperone and Transcription Factor Function. Sci Rep 2015; 5:14780. [PMID: 26437801 PMCID: PMC4594038 DOI: 10.1038/srep14780] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 09/07/2015] [Indexed: 01/24/2023] Open
Abstract
Copper (Cu), an essential micronutrient, plays a fundamental role in inflammation and angiogenesis; however, its precise mechanism remains undefined. Here we uncover a novel role of Cu transport protein Antioxidant-1 (Atox1), which is originally appreciated as a Cu chaperone and recently discovered as a Cu-dependent transcription factor, in inflammatory neovascularization. Atox1 expression is upregulated in patients and mice with critical limb ischemia. Atox1-deficient mice show impaired limb perfusion recovery with reduced arteriogenesis, angiogenesis, and recruitment of inflammatory cells. In vivo intravital microscopy, bone marrow reconstitution, and Atox1 gene transfer in Atox1−/− mice show that Atox1 in endothelial cells (ECs) is essential for neovascularization and recruitment of inflammatory cells which release VEGF and TNFα. Mechanistically, Atox1-depleted ECs demonstrate that Cu chaperone function of Atox1 mediated through Cu transporter ATP7A is required for VEGF-induced angiogenesis via activation of Cu enzyme lysyl oxidase. Moreover, Atox1 functions as a Cu-dependent transcription factor for NADPH oxidase organizer p47phox, thereby increasing ROS-NFκB-VCAM-1/ICAM-1 expression and monocyte adhesion in ECs inflamed with TNFα in an ATP7A-independent manner. These findings demonstrate a novel linkage between Atox1 and NADPH oxidase involved in inflammatory neovascularization and suggest Atox1 as a potential therapeutic target for treatment of ischemic disease.
Collapse
Affiliation(s)
- Gin-Fu Chen
- Departments of Medicine (Section of Cardiology) and Pharmacology, University of Illinois at Chicago, Chicago, IL
| | - Varadarajan Sudhahar
- Departments of Medicine (Section of Cardiology) and Pharmacology, University of Illinois at Chicago, Chicago, IL.,Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL.,Jesse Brown Veterans Affairs Medical Center, Chicago, IL
| | - Seock-Won Youn
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL.,Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL
| | - Archita Das
- Departments of Medicine (Section of Cardiology) and Pharmacology, University of Illinois at Chicago, Chicago, IL.,Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL
| | - Jaehyung Cho
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL
| | - Tetsuro Kamiya
- Departments of Medicine (Section of Cardiology) and Pharmacology, University of Illinois at Chicago, Chicago, IL.,Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL
| | - Norifumi Urao
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL
| | - Ronald D McKinney
- Departments of Medicine (Section of Cardiology) and Pharmacology, University of Illinois at Chicago, Chicago, IL.,Department of Pharmacology, University of Illinois at Chicago, Chicago, IL.,Jesse Brown Veterans Affairs Medical Center, Chicago, IL
| | - Bayasgalan Surenkhuu
- Departments of Medicine (Section of Cardiology) and Pharmacology, University of Illinois at Chicago, Chicago, IL
| | - Takao Hamakubo
- Department of Quantitative Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan
| | - Hiroko Iwanari
- Department of Quantitative Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan
| | - Senlin Li
- Department of Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - John W Christman
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine The Ohio State University Wexner Medical Center, OH
| | - Saran Shantikumar
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol
| | - Gianni D Angelini
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol.,National Heart and Lung Institute, Imperial College of London, London, UK
| | - Costanza Emanueli
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol.,National Heart and Lung Institute, Imperial College of London, London, UK
| | - Masuko Ushio-Fukai
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL.,Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL
| | - Tohru Fukai
- Departments of Medicine (Section of Cardiology) and Pharmacology, University of Illinois at Chicago, Chicago, IL.,Department of Pharmacology, University of Illinois at Chicago, Chicago, IL.,Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL.,Jesse Brown Veterans Affairs Medical Center, Chicago, IL
| |
Collapse
|
207
|
Ha YR, Kang YJ, Lee SJ. In vivo study on splenomegaly inhibition by genistein in Plasmodium berghei -infected mice. Parasitol Int 2015; 64:369-76. [DOI: 10.1016/j.parint.2015.05.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 05/13/2015] [Accepted: 05/15/2015] [Indexed: 01/24/2023]
|
208
|
Liu F, Gao X, Wang J, Gao C, Li X, Li X, Gong X, Zeng X. Transcriptome Sequencing to Identify Transcription Factor Regulatory Network and Alternative Splicing in Endothelial Cells Under VEGF Stimulation. J Mol Neurosci 2015; 58:170-7. [PMID: 26395122 DOI: 10.1007/s12031-015-0653-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 09/08/2015] [Indexed: 01/08/2023]
Abstract
This study aims to investigate the mechanisms underlying the response of human umbilical vein vascular endothelial cells (HUVECs) to vascular endothelial growth factor (VEGF) stimulation. HUVECs were treated with or without 16 ng/mL VEGF for 4 days, and RNA was extracted from HUVECs. After sequencing and data filtering (tool: NGS QC Toolkit), clean data were mapped to genome hg19 (tool: TopHat2). Thereafter, 154 differentially expressed genes (DEGs) were identified between VEGF group and control group (tool: Cuffdiff), and DEGs were enriched in 11 pathways associated with cytokine receptor interaction and chemokine signaling. Protein-protein interaction network of DEGs was constructed (tool: STRING), and ISG15 and MX1 were hub DEGs. The regulatory network of DEGs and transcription factors (TFs) (tool: TRED database) was also constructed, and CCL2 and FN1 (hub DEGs) were co-regulated by NFKB1 and RELA (hub TFs). Moreover, exon usage and alternative splicing were analyzed (tool: DEXSeq), and the splicing of ADORA2A was altered under VEGF stimulation. VEGF might influence HUVECs proliferation and migration, as well as angiogenesis process by regulating the expression of ISG15, MX1, CCL2, FN1, and ADORA2A. However, more research studies are still required to verify these predictions.
Collapse
Affiliation(s)
- Fang Liu
- Department of Neurology, China Medical University, Shenyang, 110001, China.,Department of Neurology, Center Hospital Affiliated to Shenyang Medical College, Shenyang, 110024, China
| | - Xianxin Gao
- Department of Neurology, Center Hospital Affiliated to Shenyang Medical College, Shenyang, 110024, China
| | - Jing Wang
- Department of Neurology, Center Hospital Affiliated to Shenyang Medical College, Shenyang, 110024, China
| | - Chao Gao
- Department of Neurology, Center Hospital Affiliated to Shenyang Medical College, Shenyang, 110024, China
| | - Xiaolin Li
- Department of Neurology, Center Hospital Affiliated to Shenyang Medical College, Shenyang, 110024, China
| | - Xiaodong Li
- Department of Neurology, Center Hospital Affiliated to Shenyang Medical College, Shenyang, 110024, China
| | - Xiao Gong
- Department of Neurology, Center Hospital Affiliated to Shenyang Medical College, Shenyang, 110024, China
| | - Xiandong Zeng
- The Dean's Office, Center Hospital Affiliated to Shenyang Medical College, Shenyang, 110024, China. .,China Medical University, Shenyang, 110001, China.
| |
Collapse
|
209
|
Liu RR, Li J, Gong JY, Kuang F, Liu JY, Zhang YS, Ma QL, Song CJ, Truax AD, Gao F, Yang K, Jin BQ, Chen LH. MicroRNA-141 regulates the expression level of ICAM-1 on endothelium to decrease myocardial ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol 2015; 309:H1303-13. [PMID: 26371161 DOI: 10.1152/ajpheart.00290.2015] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 09/02/2015] [Indexed: 01/01/2023]
Abstract
A growing number of studies have suggested microRNAs (miRNAs) are involved in the modulation of myocardial ischemia-reperfusion (MI/R) injury; however, the role of endogenous miRNAs targeting endothelial cells (ECs) and its interaction with ICAM-1 in the setting of MI/R remain poorly understood. Our microarray results showed that miR-146a, miR-146b-5p, miR-155*, miR-155, miR-497, and miR-451 were significantly upregulated, whereas, miR-141 and miR-564 were significantly downregulated in the ECs challenged with TNF-α for 6 h. Real-time PCR analyses additionally validated that the expression levels of miR-146a, miR-155*, and miR-141 were consistent with the microarray results. Then, ICAM-1 was identified as a novel target of miR-141 by Target Scan software and the reporter gene system. Further functional experiments showed that elevated levels of miR-141 inhibited ICAM-1 expression and diminished leukocytes adhesion to ECs in vitro. In an in vivo murine model of MI/R injury, pretreatment with miR-141 mimics through the tail vein downregulated the expression level of ICAM-1 in heart and attenuated MI/R injury as evidenced by decreased infarct size and decline of serum cardial troponin I (cTnI) and lactate dehydrogenase (LDH) concentration. The cardioprotective effects of miR-141 mimics may be attributed to the decreased infiltration of CD11b(+) cells and F4/80(+) macrophages into ischemic myocardium tissue. In conclusion, our results demonstrate that miR-141, as a novel repressor of ICAM-1, is involved in the attenuation of MI/R injury via antithetical regulation of ICAM-1 and inflammatory cells infiltration. Thus miR-141 may constitute a new therapeutic target in the setting of ischemic heart disease.
Collapse
Affiliation(s)
- Rong Rong Liu
- Department of Immunology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Jun Li
- Department of Physiology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Jiu Yu Gong
- Department of Immunology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Fang Kuang
- Department of Neurobiology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Jia Yun Liu
- Department of Clinical Laboratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China; and
| | - Yu Si Zhang
- Department of Immunology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Qian Li Ma
- Department of Immunology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Chao Jun Song
- Department of Immunology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Agnieszka D Truax
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Feng Gao
- Department of Physiology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Kun Yang
- Department of Immunology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Bo Quan Jin
- Department of Immunology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Li Hua Chen
- Department of Immunology, Fourth Military Medical University, Xi'an, People's Republic of China;
| |
Collapse
|
210
|
Exploring the various aspects of the pathological role of vascular endothelial growth factor (VEGF) in diabetic retinopathy. Pharmacol Res 2015; 99:137-48. [DOI: 10.1016/j.phrs.2015.05.013] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 05/30/2015] [Accepted: 05/31/2015] [Indexed: 12/13/2022]
|
211
|
Borgeat A, Schick C, Votta-Velis G. Pharmacologic Factors: Anaesthetic Agents that May Influence Cancer Outcomes: Local Anaesthetics. CURRENT ANESTHESIOLOGY REPORTS 2015. [DOI: 10.1007/s40140-015-0112-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
212
|
Lee W, Kim KM, Bae JS. Ameliorative Effect of Aspalathin and Nothofagin from Rooibos (Aspalathus linearis) on HMGB1-Induced Septic ResponsesIn VitroandIn Vivo. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2015. [DOI: 10.1142/s0192415x15500573] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The ubiquitous nuclear protein, high mobility group box 1 (HMGB1), is released by activated macrophages and human umbilical vein endothelial cells (HUVECs) and functions as a late mediator of experimental sepsis. Aspalathin (Asp) and nothofagin (Not), which have been reported to have anti-oxidant activity, are the two major active dihydrochalcones found in green rooibos. In this study, we investigated the antiseptic effects and underlying mechanisms of Asp and Not against HMGB1-mediated septic responses in HUVECs and mice. The anti-inflammatory activities of Asp and Not were determined by measuring permeability, monocyte adhesion and migration, and activation of proinflammatory proteins in HMGB1-activated HUVECs and mice. According to the results, Asp and Not effectively inhibited lipopolysaccharide (LPS)-induced release of HMGB1, and suppressed HMGB1-mediated septic responses, such as hyperpermeability, adhesion and migration of leukocytes, and expression of cell adhesion molecules. In addition, Asp and Not suppressed the production of tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6), the activation of nuclear factor-κB (NF-κB) and extracellular signal-regulated kinases 1 and 2 (ERK1/2) by HMGB1. Collectively, these results indicate that Asp and Not could be potential therapeutic agents for the treatment of various severe vascular inflammatory diseases via the inhibition of the HMGB1 signaling pathway.
Collapse
Affiliation(s)
- Wonhwa Lee
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biochemistry and Cell Biology, School of Medicine, Republic of Korea
| | - Kyung-Min Kim
- Division of Plant Biosciences, School of Applied BioSciences, College of Agriculture and Life Science, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Jong-Sup Bae
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Republic of Korea
| |
Collapse
|
213
|
Identification and characterization of VEGF-A-responsive neutrophils expressing CD49d, VEGFR1, and CXCR4 in mice and humans. Blood 2015; 126:2016-26. [PMID: 26286848 DOI: 10.1182/blood-2015-03-631572] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 07/23/2015] [Indexed: 12/17/2022] Open
Abstract
Vascular endothelial growth factor A (VEGF-A) is upregulated during hypoxia and is the major regulator of angiogenesis. VEGF-A expression has also been found to recruit myeloid cells to ischemic tissues where they contribute to angiogenesis. This study investigates the mechanisms underlying neutrophil recruitment to VEGF-A as well as the characteristics of these neutrophils. A previously undefined circulating subset of neutrophils shown to be CD49d(+)VEGFR1(high)CXCR4(high) was identified in mice and humans. By using chimeric mice with impaired VEGF receptor 1 (VEGFR1) or VEGFR2 signaling (Flt-1tk(-/-), tsad(-/-)), we found that parallel activation of VEGFR1 on neutrophils and VEGFR2 on endothelial cells was required for VEGF-A-induced recruitment of circulating neutrophils to tissue. Intravital microscopy of mouse microcirculation revealed that neutrophil recruitment by VEGF-A versus by the chemokine macrophage inflammatory protein 2 (MIP-2 [CXCL2]) involved the same steps of the recruitment cascade but that an additional neutrophil integrin (eg, VLA-4 [CD49d/CD29]) played a crucial role in neutrophil crawling and emigration to VEGF-A. Isolated CD49d(+) neutrophils featured increased chemokinesis but not chemotaxis compared with CD49d(-) neutrophils in the presence of VEGF-A. Finally, by targeting the integrin α4 subunit (CD49d) in a transplantation-based angiogenesis model that used avascular pancreatic islets transplanted to striated muscle, we demonstrated that inhibiting the recruitment of circulating proangiogenic neutrophils to hypoxic tissue impairs vessel neoformation. Thus, angiogenesis can be modulated by targeting cell-surface receptors specifically involved in VEGF-A-dependent recruitment of proangiogenic neutrophils without compromising recruitment of the neutrophil population involved in the immune response to pathogens.
Collapse
|
214
|
Imeri F, Blanchard O, Jenni A, Schwalm S, Wünsche C, Zivkovic A, Stark H, Pfeilschifter J, Huwiler A. FTY720 and two novel butterfly derivatives exert a general anti-inflammatory potential by reducing immune cell adhesion to endothelial cells through activation of S1P(3) and phosphoinositide 3-kinase. Naunyn Schmiedebergs Arch Pharmacol 2015; 388:1283-92. [PMID: 26267293 DOI: 10.1007/s00210-015-1159-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 07/27/2015] [Indexed: 10/23/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a key lipid regulator of a variety of cellular responses including cell proliferation and survival, cell migration, and inflammatory reactions. Here, we investigated the effect of S1P receptor activation on immune cell adhesion to endothelial cells under inflammatory conditions. We show that S1P reduces both tumor necrosis factor (TNF)-α- and lipopolysaccharide (LPS)-stimulated adhesion of Jurkat and U937 cells to an endothelial monolayer. The reducing effect of S1P was reversed by the S1P1+3 antagonist VPC23019 but not by the S1P1 antagonist W146. Additionally, knockdown of S1P3, but not S1P1, by short hairpin RNA (shRNA) abolished the reducing effect of S1P, suggesting the involvement of S1P3. A suppression of immune cell adhesion was also seen with the immunomodulatory drug FTY720 and two novel butterfly derivatives ST-968 and ST-1071. On the molecular level, S1P and all FTY720 derivatives reduced the mRNA expression of LPS- and TNF-α-induced adhesion molecules including ICAM-1, VCAM-1, E-selectin, and CD44 which was reversed by the PI3K inhibitor LY294002, but not by the MEK inhibitor U0126.In summary, our data demonstrate a novel molecular mechanism by which S1P, FTY720, and two novel butterfly derivatives acted anti-inflammatory that is by suppressing gene transcription of various endothelial adhesion molecules and thereby preventing adhesion of immune cells to endothelial cells and subsequent extravasation.
Collapse
Affiliation(s)
- Faik Imeri
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010, Bern, Switzerland
| | - Olivier Blanchard
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010, Bern, Switzerland
| | - Aurelio Jenni
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010, Bern, Switzerland
| | - Stephanie Schwalm
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010, Bern, Switzerland.,Pharmazentrum Frankfurt/ZAFES, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Christin Wünsche
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010, Bern, Switzerland.,Pharmazentrum Frankfurt/ZAFES, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Aleksandra Zivkovic
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine-University Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany
| | - Holger Stark
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine-University Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany
| | - Josef Pfeilschifter
- Pharmazentrum Frankfurt/ZAFES, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Andrea Huwiler
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010, Bern, Switzerland.
| |
Collapse
|
215
|
Yang EJ, Ku SK, Lee W, Song KS, Bae JS. Inhibitory effects of polyozellin from Polyozellus multiplex on HMGB1-mediated septic responses. Inflamm Res 2015. [PMID: 26206236 DOI: 10.1007/s00011-015-0856-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
AIM AND OBJECTIVE The ubiquitous nuclear protein, high-mobility group box 1 (HMGB1), is released by activated macrophages and human umbilical vein endothelial cells (HUVECs) and functions as a late mediator of experimental sepsis. Polyozellin, which has been reported to have a variety of biological activities including antioxidant and anticancer activity, is the major active compound found in edible mushroom (Polyozellus multiplex). In this study, we investigated the antiseptic effects and underlying mechanisms of polyozellin against HMGB1-mediated septic responses in HUVECs and mice. METHODS The anti-inflammatory activities of polyozellin were determined by measuring permeability, human neutrophil adhesion and migration, and activation of proinflammatory proteins in HMGB1-activated HUVECs and mice. RESULTS According to the results, polyozellin effectively inhibited lipopolysaccharide (LPS)-induced release of HMGB1, and suppressed HMGB1-mediated septic responses, such as hyperpermeability, adhesion and migration of leukocytes, and expression of cell adhesion molecules. In addition, polyozellin suppressed the production of tumor necrosis factor-α and interleukin (IL)-6, and the activation of nuclear factor-κB and extracellular signal-regulated kinases 1/2 by HMGB1. CONCLUSION Collectively, these results indicate that P. multiplex containing polyozellin could be commercialized as functional food for preventing and treatment of various severe vascular inflammatory diseases via inhibition of the HMGB1 signaling pathway.
Collapse
Affiliation(s)
- Eun-Ju Yang
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80 Dahak-ro, Buk-gu, Daegu, 702-701, Republic of Korea
| | | | | | | | | |
Collapse
|
216
|
Talekar M, Tran TH, Amiji M. Translational Nano-Medicines: Targeted Therapeutic Delivery for Cancer and Inflammatory Diseases. AAPS J 2015; 17:813-27. [PMID: 25921939 PMCID: PMC4477000 DOI: 10.1208/s12248-015-9772-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 04/10/2015] [Indexed: 12/12/2022] Open
Abstract
With the advent of novel and personalized therapeutic approaches for cancer and inflammatory diseases, there is a growing demand for designing delivery systems that circumvent some of the limitation with the current therapeutic strategies. Nanoparticle-based delivery of drugs has provided means of overcoming some of these limitations by ensuring the drug payload is directed to the disease site and insuring reduced off-target activity. This review highlights the challenges posed by the solid tumor microenvironment and the systemic limitations for effective chemotherapy. It then assesses the basis of nanoparticle-based targeting to the tumor tissues, which helps to overcome some of the microenvironmental and systemic limitations to therapy. We have extensively focused on some of the tumor multidrug resistance mechanisms (e.g., hypoxia and aerobic glycolysis) that contribute to the development of multidrug resistance and how targeted nano-approaches can be adopted to overcome drug resistance. Finally, we assess the combinatorial approach and how this platform has been used to develop multifunctional delivery systems for cancer therapy. The review article also focuses on inflammatory diseases, the biological therapies available for its treatment, and the concept of macrophage repolarization for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Meghna Talekar
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouve College of Health Sciences, Northeastern University, Boston, Massachusetts 02115 USA
| | - Thanh-Huyen Tran
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouve College of Health Sciences, Northeastern University, Boston, Massachusetts 02115 USA
| | - Mansoor Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Bouve College of Health Sciences, Northeastern University, Boston, Massachusetts 02115 USA
| |
Collapse
|
217
|
García-González V, Delgado-Coello B, Pérez-Torres A, Mas-Oliva J. Reality of a Vaccine in the Prevention and Treatment of Atherosclerosis. Arch Med Res 2015; 46:427-37. [PMID: 26100340 DOI: 10.1016/j.arcmed.2015.06.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 06/11/2015] [Indexed: 02/08/2023]
Abstract
Atherosclerosis together with multiple sclerosis, psoriasis and rheumatoid arthritis can be used as examples of chronic inflammatory diseases associated with multifactorial components that evolve over the years. Nevertheless, an important difference between these diseases relies on the fact that atherosclerosis develops from early ages where inflammation dominates the very beginning of the disease. This review highlights the inflammatory nature of atherosclerosis and the role the immune system plays in the process of atherogenesis. Although treatment of atherosclerosis has been for years based on lipid-lowering therapies reducing a series of risk factors, the degree of success has been only limited because cardiovascular complications related to the evolution of atherosclerotic lesions continue to appear in the population worldwide. In this sense, alternative treatments for atherosclerosis have come into play where both innate and adaptive immunity have been proposed to modulate atherosclerosis-associated inflammatory phenomena. When tested for their atheroprotective properties, several immunogens have been studied through passive and active immunization with good results and, therefore, the strategy through vaccination to control the disease has been made possible. Many experimental pre-clinical studies demonstrating proof of concept that vaccination using DNA and protein with an effective use of adjuvants and the optimal route of administration now provide a tangible new therapeutic approach that sets the stage for several of these vaccines to be tested in large, randomized, long-term clinical studies. A vaccine ready for human use will only be accomplished through the close association between academia, regulatory government organizations and private industry, allowing the reality of a simple and successful therapy to reduce atherosclerosis and its severe clinical complications.
Collapse
Affiliation(s)
- Victor García-González
- Departamento de Bioquímica y Biología Estructural, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, D.F., México
| | - Blanca Delgado-Coello
- Departamento de Bioquímica y Biología Estructural, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, D.F., México
| | - Armando Pérez-Torres
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autónoma de México, México, D.F., México
| | - Jaime Mas-Oliva
- Departamento de Bioquímica y Biología Estructural, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, D.F., México.
| |
Collapse
|
218
|
Collison JL, Carlin LM, Eichmann M, Geissmann F, Peakman M. Heterogeneity in the Locomotory Behavior of Human Monocyte Subsets over Human Vascular Endothelium In Vitro. THE JOURNAL OF IMMUNOLOGY 2015; 195:1162-70. [PMID: 26085686 DOI: 10.4049/jimmunol.1401806] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 05/22/2015] [Indexed: 12/24/2022]
Abstract
Human monocytes comprise three distinct subsets, defined by their relative expression of CD14 and CD16. These subsets appear to have different functional roles within homeostasis and inflammation, but little is known about the manner in which they interact with macro- and microvascular endothelial cells, a key enabling component for the fulfillment of their functional roles. In the present study, we examined the locomotory behavior of the three major human monocyte subsets over human endothelial monolayers subjected to physiologically relevant levels of shear flow in vitro. Each subset was shown to preferentially perform different types of locomotory behavior in a resting state. A long-range crawling behavior, similar to the "patrolling" behavior of murine Ly6C(-) monocytes, was observed in CD14(+)CD16(-) and CD14(dim)CD16(+) monocytes, but not in CD14(+)CD16(+) monocytes. CD14(dim)CD16(+) and CD14(+)CD16(+) monocytes showed a preference for adhering to microvascular over macrovascular endothelium, whereas CD14(+)CD16(-) monocytes showed the opposite. Transendothelial migration was not observed in CD14(dim)CD16(+) monocytes during the 30-min observation period. Long-range crawling behavior in CD14(dim)CD16(+) monocytes was abrogated by blockade of ICAM1, VCAM1, or CX3CL1, in contrast with CD14(+)CD16(-) monocytes, which only required ICAM1 for this behavior. These studies indicate the existence of subtype-specific human monocyte migratory behavior patterns with distinct adhesion molecule dependence, which may assist in elucidating their physiological function and relevance to disease.
Collapse
Affiliation(s)
- Joanna L Collison
- Peter Gorer Department of Immunobiology, Faculty of Life Sciences & Medicine, King's College London, London SE1 9RT, United Kingdom; and
| | - Leo M Carlin
- Centre for Molecular and Cellular Biology of Inflammation, Faculty of Life Sciences & Medicine, King's College London, London SE1 9RT, United Kingdom
| | - Martin Eichmann
- Peter Gorer Department of Immunobiology, Faculty of Life Sciences & Medicine, King's College London, London SE1 9RT, United Kingdom; and
| | - Frederic Geissmann
- Peter Gorer Department of Immunobiology, Faculty of Life Sciences & Medicine, King's College London, London SE1 9RT, United Kingdom; and Centre for Molecular and Cellular Biology of Inflammation, Faculty of Life Sciences & Medicine, King's College London, London SE1 9RT, United Kingdom
| | - Mark Peakman
- Peter Gorer Department of Immunobiology, Faculty of Life Sciences & Medicine, King's College London, London SE1 9RT, United Kingdom; and
| |
Collapse
|
219
|
Kalita J, Chauhan PS, Mani VE, Bhoi SK, Misra UK. VEGF and its receptors in dengue virus infection. J Med Virol 2015; 87:1449-55. [DOI: 10.1002/jmv.24205] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2015] [Indexed: 11/08/2022]
Affiliation(s)
- J. Kalita
- Department of Neurology; Sanjay Gandhi Postgraduate Institute of Medical Sciences; Raebareily Road Lucknow Uttar Pradesh, India
| | - P. S. Chauhan
- Department of Neurology; Sanjay Gandhi Postgraduate Institute of Medical Sciences; Raebareily Road Lucknow Uttar Pradesh, India
| | - V. E. Mani
- Department of Neurology; Sanjay Gandhi Postgraduate Institute of Medical Sciences; Raebareily Road Lucknow Uttar Pradesh, India
| | - S. K. Bhoi
- Department of Neurology; Sanjay Gandhi Postgraduate Institute of Medical Sciences; Raebareily Road Lucknow Uttar Pradesh, India
| | - U. K. Misra
- Department of Neurology; Sanjay Gandhi Postgraduate Institute of Medical Sciences; Raebareily Road Lucknow Uttar Pradesh, India
| |
Collapse
|
220
|
Lee CG, Lee HW, Kim BO, Rhee DK, Pyo S. Allicin inhibits invasion and migration of breast cancer cells through the suppression of VCAM-1: Regulation of association between p65 and ER-α. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.03.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
221
|
Abd El-Ghaffar HAER, Ahmed AI, Abdelaal AA, Emam RF, Mansour LA. Antioxidant enzymes gene polymorphisms and hepatocellular carcinoma in hepatitis C virus-infected Egyptian patients. COMPARATIVE CLINICAL PATHOLOGY 2015; 24:609-615. [DOI: 10.1007/s00580-014-1954-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
222
|
Citreoviridin Enhances Atherogenesis in Hypercholesterolemic ApoE-Deficient Mice via Upregulating Inflammation and Endothelial Dysfunction. PLoS One 2015; 10:e0125956. [PMID: 25933220 PMCID: PMC4416801 DOI: 10.1371/journal.pone.0125956] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 03/27/2015] [Indexed: 11/19/2022] Open
Abstract
Vascular endothelial dysfunction and inflammatory response are early events during initiation and progression of atherosclerosis. In vitro studies have described that CIT markedly upregulates expressions of ICAM-1 and VCAM-1 of endothelial cells, which result from NF-κB activation induced by CIT. In order to determine whether it plays a role in atherogenesis in vivo, we conducted the study to investigate the effects of CIT on atherosclerotic plaque development and inflammatory response in apolipoprotein E deficient (apoE-/-) mice. Five-week-old apoE-/- mice were fed high-fat diets and treated with CIT for 15 weeks, followed by assay of atherosclerotic lesions. Nitric oxide (NO), vascular endothelial growth factor (VEGF) and endothelin-1 (ET-1) were detected in serum. Levels of intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), VEGF, and ET-1 in plaque areas of artery walls were examined. NF-κB p65 expression and NF-κB activation in aorta also were assessed. CIT treatment significantly augmented atherosclerotic plaques and increased expressions of ICAM-1, VCAM-1, VEGF and ET-1 in aorta. Mechanistic studies showed that activation of NF-κB was significantly elevated by CIT treatment, indicating the effect of CIT on atherosclerosis may be regulated by activation of NF-κB.
Collapse
|
223
|
Kadowaki D, Anraku M, Sakaya M, Hirata S, Maruyama T, Otagiri M. Olmesartan protects endothelial cells against oxidative stress-mediated cellular injury. Clin Exp Nephrol 2015; 19:1007-14. [PMID: 25904217 DOI: 10.1007/s10157-015-1111-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 03/25/2015] [Indexed: 01/13/2023]
Abstract
BACKGROUND The primary cause of death of hemodialysis (HD) patients is cardiovascular disease, and increased oxidative stress has been proposed to be involved in the disease pathogenesis. In this study, we examined the effect of olmesartan on oxidative stress induced by angiotensin II, lipopolysaccharide, indoxyl sulfate, advanced oxidation protein products (AOPP) or hydrogen peroxide (H2O2), which are known to be present at higher concentrations in the blood of HD patients, using human umbilical vein endothelial cells (HUVECs). METHODS Oxidative stress was evaluated by measuring the mean fluorescence intensity of CM-H2DCFCA, an ROS-sensitive fluorescent dye, in HUVECs. HUVECs were incubated with each of the above compounds in the presence or absence of olmesartan. Moreover, these oxidant-stimulated cells were also treated with the reactive oxygen species (ROS) inhibitor N-acetyl-cysteine (NAC), NADPH oxidase inhibitor diphenylene iodonium (DPI) or PKC inhibitor calphostin C. In addition, we investigated the effects of olmesartan on cytotoxicity and vascular endothelial growth factor (VEGF) secretion, which is involved in vascular inflammation in HUVECs induced by AOPP or H2O2. RESULTS The treatment of these oxidant-stimulated cells with olmesartan resulted in a significant reduction in intracellular ROS production to an extent that was nearly equivalent to that of NAC, DPI or calphostin C. Furthermore, olmesartan reduced the cytotoxicity and VEGF secretion induced by AOPP or H2O2. CONCLUSIONS These results demonstrated that the antioxidant activity of olmesartan might contribute to both its vasculoprotective and anti-hypertensive effects.
Collapse
Affiliation(s)
- Daisuke Kadowaki
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
- Center for Clinical Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, Japan
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, Japan
| | - Makoto Anraku
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto, 860-0082, Japan
| | - Moe Sakaya
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
| | - Sumio Hirata
- Center for Clinical Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, Japan
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, Japan
| | - Toru Maruyama
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan.
- Center for Clinical Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, Japan.
| | - Masaki Otagiri
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto, 860-0082, Japan.
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Kumamoto, 860-0082, Japan.
| |
Collapse
|
224
|
Lee W, Yoon EK, Kim KM, Park DH, Bae JS. Antiseptic effect of vicenin-2 and scolymoside from Cyclopia subternata (honeybush) in response to HMGB1 as a late sepsis mediator in vitro and in vivo. Can J Physiol Pharmacol 2015; 93:709-20. [PMID: 26243020 DOI: 10.1139/cjpp-2015-0021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cyclopia subternata is a medicinal plant commonly used in traditional medicine to relieve pain. In this study, we investigated the antiseptic effects and underlying mechanisms of vicenin-2 and scolymoside, which are 2 active compounds from C. subternata that act against high mobility group box 1 (HMGB1)-mediated septic responses in human umbilical vein endothelial cells (HUVECs) and mice. The antiseptic activities of vicenin-2 and scolymoside were determined by measuring permeability, neutrophil adhesion and migration, and activation of proinflammatory proteins in HMGB1-activated HUVECs and mice. According to the results, vicenin-2 and scolymoside effectively inhibited lipopolysaccharide-induced release of HMGB1, and suppressed HMGB1-mediated septic responses such as hyperpermeability, the adhesion and migration of leukocytes, and the expression of cell adhesion molecules. In addition, vicenin-2 and scolymoside suppressed the production of tumor necrosis factor-α and interleukin 6, and activation of nuclear factor-κB and extracellular regulated kinases 1/2 by HMGB1. Collectively, these results indicate that vicenin-2 and scolymoside could be a potential therapeutic agents for the treatment of various severe vascular inflammatory diseases via inhibition of the HMGB1 signaling pathway.
Collapse
Affiliation(s)
- Wonhwa Lee
- a College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80 Dahak-ro, Buk-gu, Daegu 702-701, Republic of Korea.,b Department of Biochemistry and Cell Biology, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Eun-Kyung Yoon
- a College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80 Dahak-ro, Buk-gu, Daegu 702-701, Republic of Korea
| | - Kyung-Min Kim
- c Division of Plant Biosciences, School of Applied BioSciences, College of Agriculture and Life Science, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Dong Ho Park
- d Department of Ophthalmology, School of Medicine, Kyungpook National University, 50 Samduk-dong-2-ga, Jung-gu, Daegu 700-721, Republic of Korea
| | - Jong-Sup Bae
- a College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80 Dahak-ro, Buk-gu, Daegu 702-701, Republic of Korea
| |
Collapse
|
225
|
Slezak J, Kura B, Ravingerová T, Tribulova N, Okruhlicova L, Barancik M. Mechanisms of cardiac radiation injury and potential preventive approaches. Can J Physiol Pharmacol 2015; 93:737-53. [PMID: 26030720 DOI: 10.1139/cjpp-2015-0006] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
In addition to cytostatic treatment and surgery, the most common cancer treatment is gamma radiation. Despite sophisticated radiological techniques however, in addition to irradiation of the tumor, irradiation of the surrounding healthy tissue also takes place, which results in various side-effects, depending on the absorbed dose of radiation. Radiation either damages the cell DNA directly, or indirectly via the formation of oxygen radicals that in addition to the DNA damage, react with all cell organelles and interfere with their molecular mechanisms. The main features of radiation injury besides DNA damage is inflammation and increased expression of pro-inflammatory genes and cytokines. Endothelial damage and dysfunction of capillaries and small blood vessels plays a particularly important role in radiation injury. This review is focused on summarizing the currently available data concerning the mechanisms of radiation injury, as well as the effectiveness of various antioxidants, anti-inflammatory cytokines, and cytoprotective substances that may be utilized in preventing, mitigating, or treating the toxic effects of ionizing radiation on the heart.
Collapse
Affiliation(s)
- Jan Slezak
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic.,Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic
| | - Branislav Kura
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic.,Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic
| | - Táňa Ravingerová
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic.,Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic
| | - Narcisa Tribulova
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic.,Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic
| | - Ludmila Okruhlicova
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic.,Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic
| | - Miroslav Barancik
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic.,Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic
| |
Collapse
|
226
|
Levy O, Mortensen LJ, Boquet G, Tong Z, Perrault C, Benhamou B, Zhang J, Stratton T, Han E, Safaee H, Musabeyezu J, Yang Z, Multon MC, Rothblatt J, Deleuze JF, Lin CP, Karp JM. A small-molecule screen for enhanced homing of systemically infused cells. Cell Rep 2015; 10:1261-1268. [PMID: 25732817 DOI: 10.1016/j.celrep.2015.01.057] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 12/14/2014] [Accepted: 01/24/2015] [Indexed: 12/13/2022] Open
Abstract
Poor homing of systemically infused cells to disease sites may limit the success of exogenous cell-based therapy. In this study, we screened 9,000 signal-transduction modulators to identify hits that increase mesenchymal stromal cell (MSC) surface expression of homing ligands that bind to intercellular adhesion molecule 1 (ICAM-1), such as CD11a. Pretreatment of MSCs with Ro-31-8425, an identified hit from this screen, increased MSC firm adhesion to an ICAM-1-coated substrate in vitro and enabled targeted delivery of systemically administered MSCs to inflamed sites in vivo in a CD11a- (and other ICAM-1-binding domains)-dependent manner. This resulted in a heightened anti-inflammatory response. This represents a new strategy for engineering cell homing to enhance therapeutic efficacy and validates CD11a and ICAM-1 as potential targets. Altogether, this multi-step screening process may significantly improve clinical outcomes of cell-based therapies.
Collapse
Affiliation(s)
- Oren Levy
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139
- Harvard Stem Cell Institute, Cambridge, MA 02139
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139
| | - Luke J Mortensen
- Harvard Stem Cell Institute, Cambridge, MA 02139
- Wellman Center for Photomedicine and Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Gerald Boquet
- Sanofi R&D , Centre de recherche Vitry-Alfortville, 13 quai Jules Guesde, 94403 Vitry-sur-Seine Cédex, France
| | - Zhixiang Tong
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139
- Harvard Stem Cell Institute, Cambridge, MA 02139
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139
| | - Christelle Perrault
- Sanofi R&D , Centre de recherche Vitry-Alfortville, 13 quai Jules Guesde, 94403 Vitry-sur-Seine Cédex, France
| | - Brigitte Benhamou
- Sanofi R&D , Centre de recherche Vitry-Alfortville, 13 quai Jules Guesde, 94403 Vitry-sur-Seine Cédex, France
| | - Jidong Zhang
- Sanofi R&D , Centre de recherche Vitry-Alfortville, 13 quai Jules Guesde, 94403 Vitry-sur-Seine Cédex, France
| | - Tara Stratton
- Harvard Stem Cell Institute, Cambridge, MA 02139
- Wellman Center for Photomedicine and Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Edward Han
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139
- Harvard Stem Cell Institute, Cambridge, MA 02139
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139
| | - Helia Safaee
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139
- Harvard Stem Cell Institute, Cambridge, MA 02139
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139
| | - Juliet Musabeyezu
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139
- Harvard Stem Cell Institute, Cambridge, MA 02139
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139
| | - Zijiang Yang
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139
- Harvard Stem Cell Institute, Cambridge, MA 02139
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139
| | - Marie-Christine Multon
- Sanofi R&D , Centre de recherche Vitry-Alfortville, 13 quai Jules Guesde, 94403 Vitry-sur-Seine Cédex, France
| | | | - Jean-Francois Deleuze
- Sanofi R&D , Centre de recherche Vitry-Alfortville, 13 quai Jules Guesde, 94403 Vitry-sur-Seine Cédex, France
| | - Charles P Lin
- Harvard Stem Cell Institute, Cambridge, MA 02139
- Wellman Center for Photomedicine and Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Jeffrey M Karp
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02139
- Harvard Stem Cell Institute, Cambridge, MA 02139
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139
| |
Collapse
|
227
|
Li F, Zhang J, Arfuso F, Chinnathambi A, Zayed ME, Alharbi SA, Kumar AP, Ahn KS, Sethi G. NF-κB in cancer therapy. Arch Toxicol 2015; 89:711-31. [PMID: 25690730 DOI: 10.1007/s00204-015-1470-4] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 02/05/2015] [Indexed: 02/06/2023]
Abstract
The transcription factor nuclear factor kappa B (NF-κB) has attracted increasing attention in the field of cancer research from last few decades. Aberrant activation of this transcription factor is frequently encountered in a variety of solid tumors and hematological malignancies. NF-κB family members and their regulated genes have been linked to malignant transformation, tumor cell proliferation, survival, angiogenesis, invasion/metastasis, and therapeutic resistance. In this review, we highlight the diverse molecular mechanism(s) by which the NF-κB pathway is constitutively activated in different types of human cancers, and the potential role of various oncogenic genes regulated by this transcription factor in cancer development and progression. Additionally, various pharmacological approaches employed to target the deregulated NF-κB signaling pathway, and their possible therapeutic potential in cancer therapy is also discussed briefly.
Collapse
Affiliation(s)
- Feng Li
- Department of Pharmacology, Yong Loo Lin School of Medicine, Cancer Science Institute, National University of Singapore, Singapore, 117597, Singapore
| | | | | | | | | | | | | | | | | |
Collapse
|
228
|
Prakash S, Agrawal S, Kumar S, Prasad N. Impact of vascular endothelial growth factor single nucleotide polymorphism association on acute renal allograft rejection. Nephron Clin Pract 2015; 129:91-6. [PMID: 25659610 DOI: 10.1159/000368700] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 09/26/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF) is a cytokine which plays an important role in the division, proliferation and migration of endothelial cells. In the kidney, VEGF expression is found in glomerular podocytes and in tubular epithelial cells, which may result in acute inflammatory reactions. METHODS The role of VEGF gene polymorphisms (-2578 C/A, -2549 18 bp Ins/Del, -1154 G/A and +936 C/T) was investigated in 272 patients who underwent renal transplantation. ARMS-PCR and PCR-RFLP were used. Patients were categorized into acute allograft rejection (n = 76) and nonrejection (n = 196). RESULTS The VEGF -1154 GG genotype and the +936 T allele were found to be susceptible to acute rejection (AR). T-A-A-I, T-A-A-D, T-G-C-I and C-A-A-I haplotypes revealed a predisposition among AR cases. In silico analysis revealed +936 T as a significant allele involved in the transcription regulation. CONCLUSION These results highlight the role of VEGF polymorphisms in acute allograft rejection.
Collapse
Affiliation(s)
- Swayam Prakash
- Department of Medical Genetics, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | | | | | | |
Collapse
|
229
|
Sanada F, Taniyama Y, Kanbara Y, Otsu R, Ikeda-Iwabu Y, Carracedo M, Rakugi H, Morishita R. Gene therapy in peripheral artery disease. Expert Opin Biol Ther 2015; 15:381-90. [PMID: 25633211 DOI: 10.1517/14712598.2015.1007039] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Despite the remarkable progress of medicine and endovascular procedures for revascularization, patients with critical limb ischemia (CLI) remain at high risk for amputation and often have a low quality of life due to pain and ulcers in the ischemic leg. Thus, a novel strategy for generating new blood vessels in CLI patients without treatment options is vital. Pre-clinical studies and Phase I clinical trials using VEGF and fibroblast growth factor (FGF) demonstrated promising results; however, more rigorous Phase II and III clinical trials failed to demonstrate benefits for CLI patients. Recently, two multicenter, double-blind, placebo-controlled clinical trials in Japan (Phase III) and the USA (Phase II) showed the benefits of hepatocyte growth factor (HGF) gene therapy for CLI patients. Although the number of patients included in these trials was relatively small, these results imply a distinct beneficial function for HGF over other angiogenic growth factors in a clinical setting. AREAS COVERED In this review, data from Phase I-III clinical trials of gene therapy for patients with peripheral artery disease (PAD) are examined. In addition, the potential mechanisms behind the success or failure of clinical trials are discussed. EXPERT OPINION Compared with VEGF and FGF, HGF has a unique molecular effect on inflammation, fibrosis and cell senescence under pathological conditions. These features may explain the clinical benefits of HGF in PAD patients.
Collapse
Affiliation(s)
- Fumihiro Sanada
- Osaka University Graduate School of Medicine, Department of Clinical Gene Therapy , Suita, Osaka 565-0871 , Japan
| | | | | | | | | | | | | | | |
Collapse
|
230
|
Tang JM, Luo B, Xiao JH, Lv YX, Li XL, Zhao JH, Zheng F, Zhang L, Chen L, Yang JY, Guo LY, Wang L, Yan YW, Pan YM, Wang JN, Li DS, Wan Y, Chen SY. VEGF-A promotes cardiac stem cell engraftment and myocardial repair in the infarcted heart. Int J Cardiol 2015; 183:221-31. [PMID: 25679991 DOI: 10.1016/j.ijcard.2015.01.050] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 11/24/2014] [Accepted: 01/25/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND The objective of this study was to determine whether vascular endothelial growth factor (VEGF)-A subtypes improve cardiac stem cell (CSC) engraftment and promote CSC-mediated myocardial repair in the infarcted heart. METHODS CSCs were treated with VEGF receptor (VEGFR) inhibitors, VCAM-1 antibody (VCAM-1-Ab), or PKC-α inhibitor followed by the treatment with VEGF-A. CSC adhesion assays were performed in vitro. In vivo, the PKH26-labeled and VCAM-1-Ab or PKC-α inhibitor pre-treated CSCs were treated with VEGF-A followed by implantation into infarcted rat hearts. The hearts were then collected for measuring CSC engraftment and evaluating cardiac fibrosis and function 3 or 28days after the CSC transplantation. RESULTS All three VEGF-A subtypes promoted CSC adhesion to extracellular matrix and endothelial cells. VEGF-A-mediated CSC adhesion required VEGFR and PKCα signaling. Importantly, VEGF-A induced VCAM-1, but not ICAM-1 expression in CSCs through PKCα signaling. In vivo, VEGF-A promoted the engraftment of CSCs in infarcted hearts, which was attenuated by PKCα inhibitor or VCAM-1-Ab. Moreover, VEGF-A-mediated CSC engraftment resulted in a reduction in infarct size and fibrosis. Functional studies showed that the transplantation of the VEGF-A-treated CSCs stimulated extensive angiomyogenesis in infarcted hearts as indicated by the expression of cardiac troponin T and von Willebrand factor, leading to an improved performance of left ventricle. Blockade of PKCα signaling or VCAM-1 significantly diminished the beneficial effects of CSCs treated with VEGF-A. CONCLUSION VEGF-A promotes myocardial repair through, at least in part, enhancing the engraftment of CSCs mediated by PKCα/VCAM-1 pathway.
Collapse
Affiliation(s)
- Jun-Ming Tang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China; Department of Physiology and Key Lab of human Embryonic Stem Cell of Hubei Province, Hubei University of Medicine, Hubei 442000, China; Center for Medical Research and Department of Physiology, School of Basic Medical Sciences, Wuhan University, Hubei 430071, China.
| | - Bin Luo
- Department of Physiology and Key Lab of human Embryonic Stem Cell of Hubei Province, Hubei University of Medicine, Hubei 442000, China
| | - Jun-hui Xiao
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Yan-xia Lv
- Department of Physiology and Key Lab of human Embryonic Stem Cell of Hubei Province, Hubei University of Medicine, Hubei 442000, China
| | - Xiao-lin Li
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Jin-he Zhao
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Fei Zheng
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Lei Zhang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Long Chen
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Jian-Ye Yang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Lin-Yun Guo
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Lu Wang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Yu-Wen Yan
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Ya-Mo Pan
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Jia-Ning Wang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Dong-sheng Li
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Yu Wan
- Center for Medical Research and Department of Physiology, School of Basic Medical Sciences, Wuhan University, Hubei 430071, China.
| | - Shi-You Chen
- Department of Physiology & Pharmacology, The University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
231
|
Song W, Wang X. The role of TGFβ1 and LRG1 in cardiac remodelling and heart failure. Biophys Rev 2015; 7:91-104. [PMID: 28509980 PMCID: PMC4322186 DOI: 10.1007/s12551-014-0158-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 11/26/2014] [Indexed: 12/12/2022] Open
Abstract
Heart failure is a life-threatening condition that carries a considerable emotional and socio-economic burden. As a result of the global increase in the ageing population, sedentary life-style, increased prevalence of risk factors, and improved survival from cardiovascular events, the incidence of heart failure will continue to rise. Despite the advances in current cardiovascular therapies, many patients are not suitable for or may not benefit from conventional treatments. Thus, more effective therapies are required. Transforming growth factor (TGF) β family of cytokines is involved in heart development and dys-regulated TGFβ signalling is commonly associated with fibrosis, aberrant angiogenesis and accelerated progression into heart failure. Therefore, a potential therapeutic pathway is to modulate TGFβ signalling; however, broad blockage of TGFβ signalling may cause unwanted side effects due to its pivotal role in tissue homeostasis. We found that leucine-rich α-2 glycoprotein 1 (LRG1) promotes blood vessel formation via regulating the context-dependent endothelial TGFβ signalling. This review will focus on the interaction between LRG1 and TGFβ signalling, their involvement in the pathogenesis of heart failure, and the potential for LRG1 to function as a novel therapeutic target.
Collapse
Affiliation(s)
- Weihua Song
- Division of Metabolic Medicine, Lee Kong Chian School of Medicine, Nanyang Technological University, Research Techno Plaza, X-Frontiers Block, Level 4, 50 Nan yang Drive, Singapore, 637553, Singapore
| | - Xiaomeng Wang
- Division of Metabolic Medicine, Lee Kong Chian School of Medicine, Nanyang Technological University, Research Techno Plaza, X-Frontiers Block, Level 4, 50 Nan yang Drive, Singapore, 637553, Singapore. .,Division of Cell Biology in Health and Disease, Institute of Molecular and Cell Biology, Singapore Agency for Science, Technology and Research, 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore. .,Department of Cell Biology, Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK.
| |
Collapse
|
232
|
Falcinelli S, Gowen BB, Trost B, Napper S, Kusalik A, Johnson RF, Safronetz D, Prescott J, Wahl-Jensen V, Jahrling PB, Kindrachuk J. Characterization of the host response to pichinde virus infection in the Syrian golden hamster by species-specific kinome analysis. Mol Cell Proteomics 2015; 14:646-57. [PMID: 25573744 DOI: 10.1074/mcp.m114.045443] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The Syrian golden hamster has been increasingly used to study viral hemorrhagic fever (VHF) pathogenesis and countermeasure efficacy. As VHFs are a global health concern, well-characterized animal models are essential for both the development of therapeutics and vaccines as well as for increasing our understanding of the molecular events that underlie viral pathogenesis. However, the paucity of reagents or platforms that are available for studying hamsters at a molecular level limits the ability to extract biological information from this important animal model. As such, there is a need to develop platforms/technologies for characterizing host responses of hamsters at a molecular level. To this end, we developed hamster-specific kinome peptide arrays to characterize the molecular host response of the Syrian golden hamster. After validating the functionality of the arrays using immune agonists of defined signaling mechanisms (lipopolysaccharide (LPS) and tumor necrosis factor (TNF)-α), we characterized the host response in a hamster model of VHF based on Pichinde virus (PICV(1)) infection by performing temporal kinome analysis of lung tissue. Our analysis revealed key roles for vascular endothelial growth factor (VEGF), interleukin (IL) responses, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling, and Toll-like receptor (TLR) signaling in the response to PICV infection. These findings were validated through phosphorylation-specific Western blot analysis. Overall, we have demonstrated that hamster-specific kinome arrays are a robust tool for characterizing the species-specific molecular host response in a VHF model. Further, our results provide key insights into the hamster host response to PICV infection and will inform future studies with high-consequence VHF pathogens.
Collapse
Affiliation(s)
- Shane Falcinelli
- From the ‡Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Brian B Gowen
- §Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, Utah, USA
| | - Brett Trost
- ¶Department of Computer Science, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Scott Napper
- ‡‡Department of Biochemistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada, ‖Vaccine and Infectious Disease Organization-International Vaccine Center, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Anthony Kusalik
- ¶Department of Computer Science, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Reed F Johnson
- From the ‡Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - David Safronetz
- **Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Joseph Prescott
- **Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Victoria Wahl-Jensen
- §§Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, USA; ¶¶National Biodefense Analysis and Countermeasures Center, Frederick, MD 21702, USA
| | - Peter B Jahrling
- From the ‡Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA; §§Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, USA
| | - Jason Kindrachuk
- §§Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, USA;
| |
Collapse
|
233
|
Yang RC, Chang CC, Sheen JM, Wu HT, Pang JHS, Huang ST. Davallia bilabiata Inhibits TNF-α-Induced Adhesion Molecules and Chemokines by Suppressing IKK/NF-kappa B Pathway in Vascular Endothelial Cells. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2014; 42:1411-29. [DOI: 10.1142/s0192415x1450089x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Davallia bilabiata (D. bilabiata) is also called GuSuiBu in Taiwan and is used as a substitute for Drynaria fortunei J. Sm. It is often used for trauma and bone repair. The inhibitory effect of D. bilabiata on inflammatory activity has not been reported. In the present study, we aimed to study the mechanism of anti-inflammation of D. bilabiata on the adhesion of leukocytes to vascular endothelial cells. The results showed that D. bilabiata, at concentrations without cytotoxic effect, inhibited the adhesion of monocytes (THP-1) to the TNF-α-stimulated human umbilical vascular endothelial cells (HUVECs). D. bilabiata suppressed the expression of the adhesion molecules ICAM, VCAM, and E-selectin at both the mRNA and protein level. In addition, both of the TNF-α-induced mRNA and protein expression of chemokines including fractalkine/CX3CL1, MCP-1 and RANTES as well as the level of secreted soluble fractalkine were decreased by D. bilabiata. We also verified that D. bilabiata inhibited the TNF-α-induced nuclear translocation of NF-κB through the inhibitory process on the TNF-α-activated phosphorylation of IKKα, IKKβ, IκB and NF-κB. All together, we concluded that the D. bilabiata affected the canonical pathway of TNF-α-induced NF-κB activation and down-regulated cell adhesion molecules and chemokine expression through inhibition of the NF-κB/IκBα/IKK signaling pathway. These findings strongly indicated that D. bilabiata might be a promising alternative/adjunct treatment for inflammatory diseases, such as rheumatoid arthritis and osteoarthritis.
Collapse
Affiliation(s)
- Rong-Chi Yang
- Chinese Herbal Pharmacy, Chang Gung Memorial Hospital, Chang Gung University of Science and Technology, Tao-Yuan, Taiwan, ROC
| | - Cheng-Chieh Chang
- Department of Chinese Medicine and Mitochondrial Research Unit, Chang Gung University, Taiwan, ROC
| | - Jer-Ming Sheen
- Department of Chinese Medicine and Mitochondrial Research Unit, Chang Gung University, Taiwan, ROC
| | - Hsiao-Ting Wu
- Department of Chinese Medicine and Mitochondrial Research Unit, Chang Gung University, Taiwan, ROC
| | - Jong-Hwei S. Pang
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taiwan, ROC
| | - Sheng-Teng Huang
- Department of Chinese Medicine and Mitochondrial Research Unit, Chang Gung University, Taiwan, ROC
| |
Collapse
|
234
|
Tran TH, Amiji MM. Targeted delivery systems for biological therapies of inflammatory diseases. Expert Opin Drug Deliv 2014; 12:393-414. [DOI: 10.1517/17425247.2015.972931] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
235
|
Lee W, Ku SK, Lee D, Lee T, Bae JS. Emodin-6-O-β-D--glucoside inhibits high-glucose-induced vascular inflammation. Inflammation 2014; 37:306-13. [PMID: 24114447 DOI: 10.1007/s10753-013-9741-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Emodin-6-O-β-D-glucoside (EG), a new active compound from Reynoutria japonica, has recently been shown to exert potent anti-inflammatory and barrier protective effects in human umbilical vein endothelial cells (HUVECs) and in mice. Vascular inflammatory process has been suggested to play a key role in initiation and progression of atherosclerosis, a major complication of diabetes mellitus. Thus, we attempted to determine whether EG can suppress the vascular inflammatory process induced by high glucose (HG) in HUVECs and mice. Data showed that HG induced markedly increased vascular permeability, monocyte adhesion, expressions of CAMs, formation of ROS, and activation of NF-κB. Remarkably, all of the above-mentioned vascular inflammatory effects of HG were attenuated by pretreatment with EG. Vascular inflammatory responses induced by HG are critical events underlying development of various diabetic complications; therefore, our results suggest that EG may have significant therapeutic benefits against diabetic complications and atherosclerosis.
Collapse
Affiliation(s)
- Wonhwa Lee
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80 Dahak-ro, Buk-gu, Daegu, 702-701, Republic of Korea
| | | | | | | | | |
Collapse
|
236
|
Anti-septic effects of pellitorine in HMGB1-induced inflammatory responses in vitro and in vivo. Inflammation 2014; 37:338-48. [PMID: 24077682 DOI: 10.1007/s10753-013-9745-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
High mobility group box 1 (HMGB1) acts as a late mediator of vascular inflammatory conditions. Pellitorine (PT), an active amide compound from Asarum sieboldii, is known to possess antibacterial and anticancer properties. In this study, we investigated the anti-septic effects of PT against pro-inflammatory responses in human umbilical vein endothelial cells (HUVECs) induced by HMGB1 and the associated signaling pathways. According to our findings, treatment with PT resulted in inhibited release of HMGB1, down-regulation of HMGB1-dependent inflammatory responses in HUVECs, and inhibited HMGB1-mediated hyperpermeability and leukocyte migration in mice. In addition, treatment with PT resulted in reduced cecal ligation and puncture (CLP)-induced release of HMGB1 and sepsis-related mortality. PT suppressed the production of tumor necrosis factor-α and interleukin 6 and the activation of nuclear factor-κB and extracellular regulated kinases 1/2 by HMGB1. Collectively, these results indicate the potential of PT as a candidate therapeutic agent for treatment of various severe vascular inflammatory diseases via inhibition of the HMGB1 signaling pathway.
Collapse
|
237
|
Role of microRNAs in the modulation of diabetic retinopathy. Prog Retin Eye Res 2014; 43:92-107. [DOI: 10.1016/j.preteyeres.2014.07.003] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 07/22/2014] [Accepted: 07/28/2014] [Indexed: 02/02/2023]
|
238
|
Huang H, Langenkamp E, Georganaki M, Loskog A, Fuchs PF, Dieterich LC, Kreuger J, Dimberg A. VEGF suppresses T-lymphocyte infiltration in the tumor microenvironment through inhibition of NF-κB-induced endothelial activation. FASEB J 2014; 29:227-38. [PMID: 25361735 DOI: 10.1096/fj.14-250985] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Antiangiogenic treatment targeting the vascular endothelial growth factor (VEGF) signaling pathway is in clinical use, but its effect on vascular function and the tumor microenvironment is poorly understood. Here, we investigate cross-talk between VEGF and proinflammatory TNF-α signaling in endothelial cells and its impact on leukocyte recruitment. We found that cotreatment with VEGF decreased TNF-α-induced Jurkat cell adhesion to human microvascular endothelial cells by 40%. This was associated with inhibition of TNF-α-mediated regulation of 86 genes, including 2 T-lymphocyte-attracting chemokines, CXCL10 and CXCL11 [TNF-α concentration 1 ng/ml; 50% inhibition/inhibitory concentration (IC50) VEGF, 3 ng/ml]. Notably, VEGF directly suppressed TNF-α-induced gene expression through negative cross-talk with the NF-κB-signaling pathway, leading to an early decrease in IFN regulatory factor 1 (IRF-1) expression and reduced phosphorylation of signal transducer and activator of transcription 1 (p-Stat1) at later times. Inhibition of VEGF signaling in B16 melanoma tumor-bearing mice by sunitinib treatment resulted in up-regulation of CXCL10 and CXCL11 in tumor vessels, accompanied by up to 18-fold increased infiltration of CD3(+) T-lymphocytes in B16 tumors. Our results demonstrate a novel role of VEGF in negative regulation of NF-κB signaling and endothelial activation in the tumor microenvironment and provide evidence that pharmacological inhibition of VEGF signaling enhances T-lymphocyte recruitment through up-regulation of chemokines CXCL10 and CXCL11.
Collapse
Affiliation(s)
- Hua Huang
- *Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, and Department of Medical Biochemistry and Microbiology, Biomedical Center, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Elise Langenkamp
- *Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, and Department of Medical Biochemistry and Microbiology, Biomedical Center, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Maria Georganaki
- *Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, and Department of Medical Biochemistry and Microbiology, Biomedical Center, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Angelica Loskog
- *Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, and Department of Medical Biochemistry and Microbiology, Biomedical Center, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Peder Fredlund Fuchs
- *Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, and Department of Medical Biochemistry and Microbiology, Biomedical Center, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Lothar C Dieterich
- *Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, and Department of Medical Biochemistry and Microbiology, Biomedical Center, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Johan Kreuger
- *Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, and Department of Medical Biochemistry and Microbiology, Biomedical Center, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Anna Dimberg
- *Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, and Department of Medical Biochemistry and Microbiology, Biomedical Center, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
239
|
Noma H, Mimura T, Yasuda K, Shimura M. Role of inflammation in diabetic macular edema. Ophthalmologica 2014; 232:127-35. [PMID: 25342084 DOI: 10.1159/000364955] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 05/29/2014] [Indexed: 11/19/2022]
Abstract
Vitreous fluid levels of vascular endothelial growth factor (VEGF), soluble VEGF receptor (sVEGFR)-2, soluble intercellular adhesion molecule (sICAM)-1, monocyte chemotactic protein (MCP)-1 and pentraxin 3 (PTX3) were measured by enzyme-linked immunosorbent assay in 36 patients with diabetic macular edema (DME) and 15 patients with macular hole (MH). Aqueous flare values were measured with a laser flare meter, and macular edema was examined by optical coherence tomography. Vitreous fluid levels of VEGF, sVEGFR-2, sICAM-1, MCP-1 and PTX3 were significantly higher in the patients with DME than in those with MH. There was a significant correlation between the vitreous fluid level of sVEGFR-2 and the levels of sICAM-1, MCP-1 and PTX3. The aqueous flare value was significantly correlated with the vitreous fluid levels of sVEGFR-2, sICAM-1, MCP-1 and PTX3. These findings suggest that inflammatory factors may induce an increase in vascular permeability and disrupt the blood-aqueous barrier in DME patients.
Collapse
Affiliation(s)
- Hidetaka Noma
- Department of Ophthalmology, Hachioji Medical Center, Tokyo Medical University, Hachioji, Tokyo, Japan
| | | | | | | |
Collapse
|
240
|
Orosomucoid1: Involved in vascular endothelial growth factor-induced blood-brain barrier leakage after ischemic stroke in mouse. Brain Res Bull 2014; 109:88-98. [PMID: 25264156 DOI: 10.1016/j.brainresbull.2014.09.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 09/13/2014] [Accepted: 09/16/2014] [Indexed: 11/23/2022]
Abstract
Vascular endothelial growth factor (VEGF) is a promising candidate for the treatment of ischemic stroke. However, accumulating evidence demonstrated that VEGF could exacerbate blood-brain barrier (BBB) disruption after ischemic stroke. This study was designed to investigate the underlying mechanisms. In the present study, a transient (90 min) middle cerebral artery occlusion (MCAO) model was performed to induce ischemic stroke in mice. VEGF was administered intracerebroventricularly 3h after reperfusion. A gene expression microarray was utilized to investigate the differentially expressed genes among the sham, MCAO, and VEGF groups. A total of 3381 mRNAs were significantly altered by cerebral ischemia when compared with the sham group, and 15 of them were changed in the VEGF group when compared with the MCAO group. Among the 15 genes, orosomucoid (Orm) 1 was most sharply changed, and this gene has previously been reported to maintain the permeability of microvessels and integrity of the BBB. Results of the microarray showed that the expression of Orm1 increased after cerebral ischemia, whereas it decreased in response to VEGF, which was confirmed by real-time quantitative PCR, western blotting, immunohistochemistry, and immunofluorescence. The bioinformatics analysis indicated two NF-κB binding sites on the Orm1 promoter, and a super-shift assay verified that NF-κB could bind the Orm1 promoter. Results of the electrophoretic mobility shift assay (EMSA) revealed that VEGF inhibited the DNA-binding activity of NF-κB/p65. Furthermore, the elevated expression and activation of key members in the canonical NF-κB pathway induced by cerebral ischemia were also inhibited by VEGF treatment. In conclusion, this study demonstrated that decreasing the Orm1 expression via inhibition of the NF-κB pathway could be a possible mechanism involved in the aggravation of BBB disruption after stroke by VEGF.
Collapse
|
241
|
Moens S, Goveia J, Stapor PC, Cantelmo AR, Carmeliet P. The multifaceted activity of VEGF in angiogenesis - Implications for therapy responses. Cytokine Growth Factor Rev 2014; 25:473-82. [PMID: 25169850 DOI: 10.1016/j.cytogfr.2014.07.009] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 07/15/2014] [Indexed: 12/15/2022]
Abstract
Vascular endothelial growth factor (VEGF) is a key growth factor driving angiogenesis (i.e. the formation of new blood vessels) in health and disease. Pharmacological blockade of VEGF signaling to inhibit tumor angiogenesis is clinically approved but the survival benefit is limited as patients invariably acquire resistance. This is partially mediated by the intrinsic flexibility of tumor cells to adapt to VEGF-blockade. However, it has become clear that tumor stromal cells also contribute to the resistance. Originally, VEGF was thought to specifically target endothelial cells (ECs) but it is now clear that many stromal cells also respond to VEGF signaling, making anti-VEGF therapy more complex than initially anticipated. A more comprehensive understanding of the complex responses of stromal cells to VEGF-blockade might inform the design of improved anti-angiogenic agents.
Collapse
Affiliation(s)
- Stijn Moens
- Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB, K.U. Leuven, Campus Gasthuisberg, Herestraat 49, B-3000 Leuven, Belgium; Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium
| | - Jermaine Goveia
- Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB, K.U. Leuven, Campus Gasthuisberg, Herestraat 49, B-3000 Leuven, Belgium; Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium
| | - Peter C Stapor
- Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB, K.U. Leuven, Campus Gasthuisberg, Herestraat 49, B-3000 Leuven, Belgium; Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium
| | - Anna Rita Cantelmo
- Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB, K.U. Leuven, Campus Gasthuisberg, Herestraat 49, B-3000 Leuven, Belgium; Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB, K.U. Leuven, Campus Gasthuisberg, Herestraat 49, B-3000 Leuven, Belgium; Laboratory of Angiogenesis & Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium.
| |
Collapse
|
242
|
Nuclear factor kappa-B signaling is integral to ocular neovascularization in ischemia-independent microenvironment. PLoS One 2014; 9:e101602. [PMID: 25050547 PMCID: PMC4106825 DOI: 10.1371/journal.pone.0101602] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 06/09/2014] [Indexed: 01/24/2023] Open
Abstract
Retinal ischemia promotes the upregulation of VEGF expression and accounts for most pathological features of retinal neovascularization (NV). Paradoxically, VEGF remains the pivotal stimulator of ocular NV, despite the absence of ischemia. Therefore, the central question arises as to how the various molecular mechanisms interplay in ischemia-independent NV. It's been suggested that NFκB plays a crucial role in the pathogenesis of diabetic vasculopathies. Here, we dissected the molecular mechanism of ocular NV in the rho/VEGF transgenic mouse model, which develops subretinal NV in ischemia-independent microenvironment. Furthermore, we examined whether intravitreal administration of YC-1, a HIF-1 inhibitor, can modulate the activation of NFκB and its downstream angiogenic signaling in the mouse retina. We demonstrated that YC-1 inhibited retinal NFκB/p65 DNA binding activity and downregulated NFκB/p65, FAK, α5β1, EPO, ET-1, and MMP-9 expression at the message and the protein levels. In addition, YC-1 significantly inhibited subretinal NV by reducing the number of neovascular lesions, the area of each lesion and the total area of NV per retina. We further investigated the influence of VEGF signaling pathway on HIF-1α transcriptional activity to substantiate that this mouse model develops subretinal NV in an ischemia-independent microenvironment. Our data demonstrated that VEGF overexpression didn't have any impact on HIF-1α transcriptional activity, whereas treatment with YC-1 significantly inhibited endogenous HIF-1 activity. Our study suggests that retinal NFκB transcriptional activity is pivotal to ischemia-independent mechanisms, which lead to the local activation of angiogenic cascades. Our data also indicate that the nexus between VEGF and NFκB is implicated in triggering the angiogenic cascade that promotes retinal NV. Hence, targeting the VEGF/NFκB axis may act in a negative feedback loop to suppress ocular NV. This study suggests that inhibition of NFκB activation may be a means of turning off a “master switch” responsible for initiating and perpetuating these ocular pathologies.
Collapse
|
243
|
Nassar ZD, Moon H, Duong T, Neo L, Hill MM, Francois M, Parton RG, Parat MO. PTRF/Cavin-1 decreases prostate cancer angiogenesis and lymphangiogenesis. Oncotarget 2014; 4:1844-55. [PMID: 24123650 PMCID: PMC3858569 DOI: 10.18632/oncotarget.1300] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Caveolae are specialized plasma membrane subdomains implicated in cellular functions such as migration, signalling and trafficking. Caveolin-1 and polymerase I and transcript release factor (PTRF)/cavin-1 are essential for caveola formation. Caveolin-1 is overexpressed and secreted in prostate tumors and promotes aggressiveness and angiogenesis. In contrast, a lack of PTRF expression is reported in prostate cancer, and ectopic PTRF expression in prostate cancer cells inhibits tumor growth and metastasis. We experimentally manipulated PTRF expression in three prostate cancer cell lines, namely the caveolin-1 positive cells PC3 and DU145 and the caveolin-1-negative LNCaP cells, to evaluate angiogenesis- and lymphangiogenesis-regulating functions of PTRF. We show that the conditioned medium of PTRF-expressing prostate cancer cells decreases ECs proliferation, migration and differentiation in vitro and ex vivo. This can occur independently from caveolin-1 expression and secretion or caveola formation, since the anti-angiogenic effects of PTRF were detected in caveolin-1-negative LNCaP cells. Additionally, PTRF expression in PC3 cells significantly decreased blood and lymphatic vessel densities in orthotopic tumors in mice. Our results suggest that the absence of PTRF in prostate cancer cells contributes significantly to tumour progression and metastasis by promoting the angiogenesis and lymphangiogenesis potential of the cancer cells, and this could be exploited for therapy.
Collapse
Affiliation(s)
- Zeyad D Nassar
- The University of Queensland, School of Pharmacy, QLD, Australia
| | | | | | | | | | | | | | | |
Collapse
|
244
|
Wang F, Liu S, Sun Q, Zhang L, Song Y, Sheng W, Xi S, Sun G. Urinary VEGF and PGE2 levels and the association with arsenical metabolites in copper-smelting workers. Occup Environ Med 2014; 71:675-80. [PMID: 24996682 DOI: 10.1136/oemed-2014-102173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVES To examine vascular endothelial growth factor (VEGF) and PGE2 levels in urine from the copper smelting workers exposed to arsenic and analyse the relationships between urinary VEGF or PGE2 level and arsenical metabolites. METHODS The study was conducted in a group of 106 copper-smelting male workers. Information about each subject was obtained by questionnaire, inorganic As (iAs), monomethylarsonic acid (MMA), dimethylarsinic acid (DMA), VEGF and prostaglandin E2 (PGE2) in urine were determined. Standing height, body weight, and blood pressure were measured. RESULTS According to the urine arsenic levels, participants were separated into three groups: Group 1: urine total arsenic <35 mg/L, Group 2: 35-100 mg/L, and Group 3: >100 mg/L. The median levels of urinary VEGF and PGE2 in Groups 1, 2 and 3 were 10.57 and 1032.0 pg/mL, 24.39 and 1060.9 pg/mL, and 49.0 and 1330.4 pg/mL, respectively. Urinary VEGF levels were positive associated with arsenical metabolites (iAs, MMA, DMA and TAs). Additionally, urinary VEGF and PGE2 levels were all correlated positively with the urinary MMA% (r=0.221, p=0.026 and r=0.206, p=0.037). While urinary VEGF was negatively with DMA% and secondary methylation index (r=-0.242, p=0.014 and r=-0.214, p=0.030, respectively). CONCLUSIONS Urinary VEGF and PGE2 levels increased in arsenic exposure copper smelting workers, and urinary VEGF levels are well associated with the urinary arsenicals. This finding may provide useful information for developing measurement, prevention and treatment of damage induced by arsenic in the future.
Collapse
Affiliation(s)
- Fei Wang
- Department of Occupational and Environmental Health, Liaoning Provincial Key Laboratory of Arsenic Biological Effect and Poisoning, School of Public Health, China Medical University, Shenyang, China
| | - Shengnan Liu
- Department of Occupational and Environmental Health, Liaoning Provincial Key Laboratory of Arsenic Biological Effect and Poisoning, School of Public Health, China Medical University, Shenyang, China
| | - Qingshan Sun
- Department of Occupational and Environmental Health, Liaoning Provincial Key Laboratory of Arsenic Biological Effect and Poisoning, School of Public Health, China Medical University, Shenyang, China
| | - Lin Zhang
- Department of Occupational and Environmental Health, Liaoning Provincial Key Laboratory of Arsenic Biological Effect and Poisoning, School of Public Health, China Medical University, Shenyang, China
| | - Yingli Song
- Department of Occupational and Environmental Health, Liaoning Provincial Key Laboratory of Arsenic Biological Effect and Poisoning, School of Public Health, China Medical University, Shenyang, China
| | - Wei Sheng
- Shenyang Health Inspection Bureau, Shenyang City, China
| | - Shuhua Xi
- Department of Occupational and Environmental Health, Liaoning Provincial Key Laboratory of Arsenic Biological Effect and Poisoning, School of Public Health, China Medical University, Shenyang, China
| | - Guifan Sun
- Department of Occupational and Environmental Health, Liaoning Provincial Key Laboratory of Arsenic Biological Effect and Poisoning, School of Public Health, China Medical University, Shenyang, China
| |
Collapse
|
245
|
Vascular endothelial growth factor (VEGF) and inducible nitric oxide synthase (iNOS) in oral lichen planus: An immunohistochemical study for the correlation between vascular and inflammatory reactions. JOURNAL OF ORAL AND MAXILLOFACIAL SURGERY MEDICINE AND PATHOLOGY 2014. [DOI: 10.1016/j.ajoms.2013.09.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
246
|
Fearnley GW, Odell AF, Latham AM, Mughal NA, Bruns AF, Burgoyne NJ, Homer-Vanniasinkam S, Zachary IC, Hollstein MC, Wheatcroft SB, Ponnambalam S. VEGF-A isoforms differentially regulate ATF-2-dependent VCAM-1 gene expression and endothelial-leukocyte interactions. Mol Biol Cell 2014; 25:2509-21. [PMID: 24966171 PMCID: PMC4142621 DOI: 10.1091/mbc.e14-05-0962] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
VEGF-A isoforms differentially stimulate endothelial VCAM-1 gene expression via an ERK1/2 protein kinase and ATF-2 transcription factor–dependent mechanism. Such signal transduction enables VEGF-A isoform–specific stimulation of leukocyte binding to endothelial cells, explaining how inflammation could be differentially regulated. Vascular endothelial growth factor A (VEGF-A) regulates many aspects of vascular physiology. VEGF-A stimulates signal transduction pathways that modulate endothelial outputs such as cell migration, proliferation, tubulogenesis, and cell–cell interactions. Multiple VEGF-A isoforms exist, but the biological significance of this is unclear. Here we analyzed VEGF-A isoform–specific stimulation of VCAM-1 gene expression, which controls endothelial–leukocyte interactions, and show that this is dependent on both ERK1/2 and activating transcription factor-2 (ATF-2). VEGF-A isoforms showed differential ERK1/2 and p38 MAPK phosphorylation kinetics. A key feature of VEGF-A isoform–specific ERK1/2 activation and nuclear translocation was increased phosphorylation of ATF-2 on threonine residue 71 (T71). Using reverse genetics, we showed ATF-2 to be functionally required for VEGF-A–stimulated endothelial VCAM-1 gene expression. ATF-2 knockdown blocked VEGF-A–stimulated VCAM-1 expression and endothelial–leukocyte interactions. ATF-2 was also required for other endothelial cell outputs, such as cell migration and tubulogenesis. In contrast, VCAM-1 was essential only for promoting endothelial–leukocyte interactions. This work presents a new paradigm for understanding how soluble growth factor isoforms program complex cellular outputs and responses by modulating signal transduction pathways.
Collapse
Affiliation(s)
- Gareth W Fearnley
- Endothelial Cell Biology Unit, School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Adam F Odell
- Endothelial Cell Biology Unit, School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Antony M Latham
- Endothelial Cell Biology Unit, School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Nadeem A Mughal
- Endothelial Cell Biology Unit, School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, United KingdomLeeds Vascular Institute, Leeds General Infirmary, Leeds LS1 3EX, United Kingdom
| | - Alexander F Bruns
- Division of Cardiovascular and Diabetes Research, Faculty of Medicine and Health, LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom
| | | | | | - Ian C Zachary
- Division of Cardiovascular Biology and Medicine, Rayne Institute, University College London, London, United Kingdom
| | | | - Stephen B Wheatcroft
- Division of Cardiovascular and Diabetes Research, Faculty of Medicine and Health, LIGHT Laboratories, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Sreenivasan Ponnambalam
- Endothelial Cell Biology Unit, School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
247
|
Kwak S, Ku SK, Bae JS. Fisetin inhibits high-glucose-induced vascular inflammation in vitro and in vivo. Inflamm Res 2014; 63:779-87. [PMID: 24923846 DOI: 10.1007/s00011-014-0750-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 05/08/2014] [Accepted: 05/30/2014] [Indexed: 11/28/2022] Open
Abstract
AIM AND OBJECTIVE Fisetin, an active compound isolated from flowering plants in the family Fabaceae, was reported to have antiviral, neuroprotective, and anti-inflammatory effects. Vascular inflammatory processes have been suggested to play key roles in the initiation and progression of atherosclerosis, a major complication of diabetes mellitus. Thus, we determined the ability of fisetin to suppress vascular inflammatory processes induced by high glucose (HG) in primary human umbilical vein endothelial cells (HUVECs) and mice. METHODS The effects of fisetin on HG-induced vascular inflammation were determined by measuring vascular permeability, leukocyte adhesion and migration, cell adhesion molecule (CAM) expression levels, reactive oxygen species (ROS) formation, and nuclear factor (NF)-κB activation. RESULTS HG markedly increased vascular permeability, monocyte adhesion, expressions of CAMs, formation of ROS, and activation of NF-κB. Remarkably, all of the observed vascular inflammatory effects induced by HG were inhibited by pretreatment with fisetin. CONCLUSION Vascular inflammatory responses induced by HG are critical events underlying the development of diabetic complications; therefore, our results suggest that fisetin possesses significant therapeutic effects against diabetic complications and atherosclerosis.
Collapse
Affiliation(s)
- Soyoung Kwak
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, 80 Dahak-ro Buk-gu, Daegu, 702-701, Republic of Korea
| | | | | |
Collapse
|
248
|
Gambogic acid exhibits anti-psoriatic efficacy through inhibition of angiogenesis and inflammation. J Dermatol Sci 2014; 74:242-50. [DOI: 10.1016/j.jdermsci.2014.03.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 01/29/2014] [Accepted: 03/03/2014] [Indexed: 11/24/2022]
|
249
|
Schlesinger M, Bendas G. Vascular cell adhesion molecule-1 (VCAM-1)--an increasing insight into its role in tumorigenicity and metastasis. Int J Cancer 2014; 136:2504-14. [PMID: 24771582 DOI: 10.1002/ijc.28927] [Citation(s) in RCA: 161] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Accepted: 04/16/2014] [Indexed: 12/14/2022]
Abstract
Vascular cell adhesion molecule-1 (VCAM-1) first attracted attention more than two decades ago as endothelial adhesion receptor with key function for leukocyte recruitment in term of cellular immune response. The early finding of VCAM-1 binding to melanoma cells, and thus a suggested mechanistic contribution to metastatic spread, was the first and for a long time the only link of VCAM-1 to cancer sciences. In the last few years, hallmarked by a growing insight into the molecular understanding of tumorigenicity and metastasis, an impressive variety of VCAM-1 functionalities in cancer have been elucidated. The present review aims to provide a current overview of VCAM-1 relevance for tumor growth, metastasis, angiogenesis, and related processes. By illustrating the intriguing role of VCAM-1 in cancer disease, VCAM-1 is suggested as a new and up to now underestimated target in cancer treatment and in clinical diagnosis of malignancies.
Collapse
Affiliation(s)
- Martin Schlesinger
- Department of Pharmacy, Rheinische Friedrich-Wilhelms-University Bonn, 53121, Bonn, Germany
| | | |
Collapse
|
250
|
Marra A, Vargas M, Striano P, Del Guercio L, Buonanno P, Servillo G. Posterior reversible encephalopathy syndrome: The endothelial hypotheses. Med Hypotheses 2014; 82:619-22. [DOI: 10.1016/j.mehy.2014.02.022] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Revised: 02/13/2014] [Accepted: 02/21/2014] [Indexed: 01/01/2023]
|