Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(3)2022 Jan 24.
Article in English | MEDLINE | ID: mdl-35163238

ABSTRACT

Blood fluid shear stress (FSS) modulates endothelial function and vascular pathophysiology. The small extracellular vesicles (sEVs) such as exosomes are potent mediators of intercellular communication, and their contents reflect cellular stress. Here, we explored the miRNA profiles in endothelial cells (EC)-derived sEVs (EC-sEVs) under atheroprotective laminar shear stress (LSS) and atheroprone low-oscillatory shear stress (OSS) and conducted a network analysis to identify the main biological processes modulated by sEVs' miRNAs. The EC-sEVs were collected from culture media of human umbilical vein endothelial cells exposed to atheroprotective LSS (20 dyne/cm2) and atheroprone OSS (±5 dyne/cm2). We explored the miRNA profiles in FSS-induced EC-sEVs (LSS-sEVs and OSS-sEVs) and conducted a network analysis to identify the main biological processes modulated by sEVs' miRNAs. In vivo studies were performed in a mouse model of partial carotid ligation. The sEVs' miRNAs-targeted genes were enriched for endothelial activation such as angiogenesis, cell migration, and vascular inflammation. OSS-sEVs promoted tube formation, cell migration, monocyte adhesion, and apoptosis, and upregulated the expression of proteins that stimulate these biological processes. FSS-induced EC-sEVs had the same effects on endothelial mechanotransduction signaling as direct stimulation by FSS. In vivo studies showed that LSS-sEVs reduced the expression of pro-inflammatory genes, whereas OSS-sEVs had the opposite effect. Understanding the landscape of EC-exosomal miRNAs regulated by differential FSS patterns, this research establishes their biological functions on a system level and provides a platform for modulating the overall phenotypic effects of sEVs.


Subject(s)
Endothelial Cells/physiology , Extracellular Vesicles/genetics , Mechanotransduction, Cellular/physiology , Animals , Apoptosis/genetics , Cell Movement/genetics , Cells, Cultured , Extracellular Vesicles/metabolism , Gene Expression/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Signal Transduction/genetics , Stress, Mechanical , Transcriptome/genetics
2.
Int J Mol Sci ; 22(13)2021 Jul 01.
Article in English | MEDLINE | ID: mdl-34281156

ABSTRACT

Cardiotoxicity is associated with the long-term clinical application of doxorubicin (DOX) in cancer patients. Mesenchymal stem cell-derived small extracellular vesicles (MSC-sEVs) including exosomes have been suggested for the treatment of various diseases, including ischemic diseases. However, the effects and functional mechanism of MSC-sEVs in DOX-induced cardiomyopathy have not been clarified. Here, MSC-sEVs were isolated from murine embryonic mesenchymal progenitor cell (C3H/10T1/2) culture media, using ultrafiltration. H9c2 cardiac myoblast cells were pretreated with MSC-sEVs and then exposed to DOX. For in vivo studies, male C57BL/6 mice were administered MSC-sEVs intravenously, prior to a single dose of DOX (15 mg/kg, intraperitoneal). The mice were sacrificed 14 days after DOX treatment. The results showed that MSC-sEVs protected cardiomyocytes from DOX-induced cell death. H9c2 cells treated with DOX showed downregulation of both phosphorylated Akt and survivin, whereas the treatment of MSC-sEVs recovered expression, indicating their anti-apoptotic effects. Three microRNAs (miRNAs) (miR 199a-3p, miR 424-5p, and miR 21-5p) in MSC-sEVs regulated the Akt-Sp1/p53 signaling pathway in cardiomyocytes. Among them, miR 199a-3p was involved in regulating survivin expression, which correlated with the anti-apoptotic effects of MSC-sEVs. In in vivo studies, the echocardiographic results showed that the group treated with MSC-sEVs recovered from DOX-induced cardiomyopathy, showing improvement of both the left ventricle fraction and ejection fraction. MSC-sEVs treatment also increased both survivin and B-cell lymphoma 2 expression in heart tissue compared to the DOX group. Our results demonstrate that MSC-sEVs have protective effects against DOX-induced cardiomyopathy by upregulating survivin expression, which is mediated by the regulation of Akt activation by miRNAs in MSC-sEVs. Thus, MSC-sEVs may be a novel therapy for the prevention of DOX-induced cardiomyopathy.


Subject(s)
Cardiomyopathies/metabolism , Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/metabolism , Animals , Apoptosis/drug effects , Cardiomyopathies/prevention & control , Cardiotoxicity/metabolism , Doxorubicin/adverse effects , Doxorubicin/pharmacology , Exosomes/metabolism , Extracellular Vesicles/physiology , Male , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction/drug effects , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Survivin/genetics , Survivin/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
3.
Exp Mol Med ; 51(11): 1-15, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31776326

ABSTRACT

Endothelial mechanotransduction by fluid shear stress (FSS) modulates endothelial function and vascular pathophysiology through mechanosensors on the cell membrane. The coxsackievirus and adenovirus receptor (CAR) is not only a viral receptor but also a component of tight junctions and plays an important role in tissue homeostasis. Here, we demonstrate the expression, regulatory mechanism, and role of CAR in vascular endothelial cells (ECs) under FSS conditions. Disturbed flow increased, whereas unidirectional laminar shear stress (LSS) decreased, CAR expression in ECs through the Krüppel-like factor 2 (KLF2)/activator protein 1 (AP-1) axis. Deletion of CAR reduced the expression of proinflammatory genes and endothelial inflammation induced by disturbed flow via the suppression of NF-κB activation. Consistently, disturbed flow-induced atherosclerosis was reduced in EC-specific CAR KO mice. CAR was found to be involved in endothelial mechanotransduction through the regulation of platelet endothelial cell adhesion molecule 1 (PECAM-1) phosphorylation. Our results demonstrate that endothelial CAR is regulated by FSS and that this regulated CAR acts as an important modulator of endothelial mechanotransduction by FSS.


Subject(s)
Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Shear Strength/physiology , Animals , Atherosclerosis/genetics , Atherosclerosis/metabolism , Blotting, Western , Cell Line , Coxsackie and Adenovirus Receptor-Like Membrane Protein/genetics , Fluorescent Antibody Technique , Human Umbilical Vein Endothelial Cells , Humans , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/genetics , Phosphorylation/physiology , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology , Stress, Mechanical , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
4.
Sci Rep ; 9(1): 12149, 2019 08 21.
Article in English | MEDLINE | ID: mdl-31434946

ABSTRACT

Doxorubicin (DOX) is a widely used anti-cancer drug; however, it has limited application due to cardiotoxicity. Extracorporeal shock waves (ESW) have been suggested to treat inflammatory and ischemic diseases, but the concrete effect of ESW in DOX-induced cardiomyopathy remain obscure. After H9c2 cells were subjected to ESW (0.04 mJ/cm2), they were treated with 1 µM DOX. As a result, ESW protected cardiomyocytes from DOX-induced cell death. H9c2 cells treated with DOX downregulated p-Akt and survivin expression, whereas the ESW treatment recovered both, suggesting its anti-apoptotic effect. ESW activated integrin αvß3 and αvß5, cardiomyocyte mechanosensors, followed by upregulation of ILK, p-Akt and survivin levels. Further, Sp1 and p53 were determined as key transcriptional factors mediating survivin expression via Akt phosphorylation by ESW. In in vivo acute DOX-induced cardiomyopathy model, the echocardiographic results showed that group subjected to ESW recovered from acute DOX-induced cardiomyopathy; left ventricular function was improved. The immunohistochemical staining results showed increased survivin and Bcl2 expression in ESW + DOX group compared to those in the DOX-injected group. In conclusion, non-invasive shockwaves protect cardiomyocytes from DOX-induced cardiomyopathy by upregulating survivin via integrin-ILK-Akt-Sp1/p53 pathway. In vivo study proposed ESW as a new kind of specific and safe therapy against acute DOX-induced cardiomyopathy.


Subject(s)
High-Energy Shock Waves , Myocytes, Cardiac/metabolism , Signal Transduction/radiation effects , Survivin/metabolism , Up-Regulation/radiation effects , Animals , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Cardiomyopathies/chemically induced , Cardiomyopathies/pathology , Cardiomyopathies/therapy , Cell Line , Doxorubicin/toxicity , Integrins/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Sp1 Transcription Factor/metabolism , Survivin/antagonists & inhibitors , Survivin/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation/drug effects
5.
Atherosclerosis ; 270: 76-88, 2018 03.
Article in English | MEDLINE | ID: mdl-29407891

ABSTRACT

BACKGROUND AND AIMS: Vascular endothelial cells (ECs) are exposed to fluid shear stress (FSS), which modulates vascular pathophysiology. Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is crucial in endothelial dysfunction and atherosclerosis. We elucidated the mechanism regulating LOX-1 expression in ECs by FSS. METHODS: Human umbilical vein endothelial cells were exposed to laminar shear stress (LSS) of indicated intensities using a unidirectional steady flow, or to oscillatory shear stress (OSS) using a bidirectional disturbed flow. In vivo studies were performed in a mouse model of partial carotid ligation and human pulmonary artery sections. RESULTS: Within ECs, OSS upregulated LOX-1 expression, while LSS (20 dyne/cm2) downregulated it. We confirmed that OSS-induced LOX-1 expression was suppressed when the mechanotransduction was inhibited by knockdown of the mechanosensory complex. In addition, we demonstrated that Kruppel-like factor 2 (KLF2) has an inhibitory role on OSS-induced LOX-1 expression. Next, we determined that activator protein-1 (AP-1) was the key transcription factor inducing LOX-1 expression by OSS, which was inhibited by KLF2 overexpression. To explore whether the intensity of LSS affects LOX-1 expression, we tested three different intensities (20, 60, and 120 dyne/cm2) of LSS. We observed higher LOX-1 expression with high shear stresses of 120 dyne/cm2 compared to 20 and 60 dyne/cm2, with OSS-like KLF2-AP-1 signaling patterns. Furthermore, ECs within disturbed flow regions showed upregulated LOX-1 expression in vivo. CONCLUSIONS: We concluded that LOX-1 expression on ECs is regulated via FSS depending on its intensity as well as pattern. Furthermore, this is mediated through the KLF2-AP1 pathway of mechanotransduction.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , Kruppel-Like Transcription Factors/metabolism , Mechanotransduction, Cellular , Scavenger Receptors, Class E/metabolism , Transcription Factor AP-1/metabolism , Animals , Carotid Stenosis/genetics , Carotid Stenosis/metabolism , Carotid Stenosis/pathology , Cells, Cultured , Disease Models, Animal , Human Umbilical Vein Endothelial Cells/pathology , Humans , Kruppel-Like Transcription Factors/genetics , Lipoproteins, LDL/metabolism , Male , Mice, Inbred C57BL , Regional Blood Flow , Scavenger Receptors, Class E/genetics , Stress, Mechanical , Transcription Factor AP-1/genetics , Up-Regulation
6.
Sci Rep ; 6: 25636, 2016 05 12.
Article in English | MEDLINE | ID: mdl-27173134

ABSTRACT

Atherosclerosis occurs preferentially in arterial regions exposed to disturbed blood flow. Targeting these pro-atherogenic regions is a potential anti-atherogenic therapeutic approach, but it has been extremely challenging. Here, using in vivo phage display approach and the partial carotid ligation model of flow-induced atherosclerosis in mouse, we identified novel peptides that specifically bind to endothelial cells (ECs) exposed to disturbed flow condition in pro-atherogenic regions. Two peptides, CLIRRTSIC and CPRRSHPIC, selectively bound to arterial ECs exposed to disturbed flow not only in the partially ligated carotids but also in the lesser curvature and branching point of the aortic arch in mice as well as human pulmonary artery branches. Peptides were conjugated to branched polyethylenimine-polyethylene glycol polymer to generate polyplexes carrying siRNA targeting intercellular adhesion molecule-1 (siICAM-1). In mouse model, CLIRRTSIC polyplexes carrying si-ICAM-1 specifically bound to endothelium in disturbed flow regions, reducing endothelial ICAM-1 expression. Mass spectrometry analysis revealed that non-muscle myosin heavy chain II A (NMHC IIA) is a protein targeted by CLIRRTSIC peptide. Further studies showed that shear stress regulates NMHC IIA expression and localization in ECs. The CLIRRTSIC is a novel peptide that could be used for targeted delivery of therapeutics such as siRNAs to pro-atherogenic endothelium.


Subject(s)
Atherosclerosis/prevention & control , Endothelium, Vascular/drug effects , Peptides/pharmacology , RNA, Small Interfering/genetics , Amino Acid Sequence , Animals , Atherosclerosis/genetics , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Gene Expression/drug effects , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Male , Mice, Inbred C57BL , Myosin Heavy Chains/metabolism , Nonmuscle Myosin Type IIA/metabolism , Peptides/metabolism , Protein Binding
7.
PLoS One ; 11(1): e0147839, 2016.
Article in English | MEDLINE | ID: mdl-26807573

ABSTRACT

A naturally occurring bile acid, ursodeoxycholic acid (UDCA), is known to alleviate endoplasmic reticulum (ER) stress at the cellular level. However, the detailed action mechanisms of UDCA in atherosclerosis are not fully understood. In this study, we demonstrated whether UDCA exerts anti-atherogenic activity in diabetic atherosclerosis by targeting ER stress and "receptor for advanced glycation endproduct" (RAGE) signaling. UDCA markedly reduced ER stress, RAGE expression, and pro-inflammatory responses [including NF-κB activation and reactive oxygen species (ROS) production] induced in endothelial cells (ECs) by high glucose (HG). In particular, UDCA inhibited HG-induced ROS production by increasing the Nrf2 level. In macrophages, UDCA also blocked HG-induced RAGE and pro-inflammatory cytokine expression and inhibited foam cell formation via upregulation of the ATP-binding cassette (ABC) transporters, ABCA1 and ABCG1. In the diabetic mouse model, UDCA inhibited atheromatous plaque formation by decreasing ER stress, and the levels of RAGE and adhesion molecules. In conclusion, UDCA exerts an anti-atherogenic activity in diabetic atherosclerosis by targeting both ER stress and RAGE signaling. Our work implicates UDCA as a potential therapeutic agent for prevention or treatment of diabetic atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetic Angiopathies/metabolism , Receptor for Advanced Glycation End Products/metabolism , Signal Transduction/drug effects , Ursodeoxycholic Acid/pharmacology , Animals , Endoplasmic Reticulum Stress/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Mice , NF-kappa B/metabolism , Oxidative Stress/drug effects , Plaque, Atherosclerotic/metabolism , Reactive Oxygen Species/metabolism
8.
Mol Cells ; 38(10): 851-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26442866

ABSTRACT

Disturbed blood flow with low-oscillatory shear stress (OSS) is a predominant atherogenic factor leading to dysfunctional endothelial cells (ECs). Recently, it was found that disturbed flow can directly induce endoplasmic reticulum (ER) stress in ECs, thereby playing a critical role in the development and progression of atherosclerosis. Ursodeoxycholic acid (UDCA), a naturally occurring bile acid, has long been used to treat chronic cholestatic liver disease and is known to alleviate endoplasmic reticulum (ER) stress at the cellular level. However, its role in atherosclerosis remains unexplored. In this study, we demonstrated the anti-atherogenic activity of UDCA via inhibition of disturbed flow-induced ER stress in atherosclerosis. UDCA effectively reduced ER stress, resulting in a reduction in expression of X-box binding protein-1 (XBP-1) and CEBP-homologous protein (CHOP) in ECs. UDCA also inhibits the disturbed flow-induced inflammatory responses such as increases in adhesion molecules, monocyte adhesion to ECs, and apoptosis of ECs. In a mouse model of disturbed flow-induced atherosclerosis, UDCA inhibits atheromatous plaque formation through the alleviation of ER stress and a decrease in adhesion molecules. Taken together, our results revealed that UDCA exerts anti-atherogenic activity in disturbed flow-induced atherosclerosis by inhibiting ER stress and the inflammatory response. This study suggests that UDCA may be a therapeutic agent for prevention or treatment of atherosclerosis.


Subject(s)
Atherosclerosis/prevention & control , Endoplasmic Reticulum Stress/drug effects , Plaque, Atherosclerotic/prevention & control , Ursodeoxycholic Acid/pharmacology , Animals , Apoptosis/drug effects , Atherosclerosis/etiology , Atherosclerosis/metabolism , Blood Circulation , Carotid Artery, Common/physiopathology , Cell Adhesion/drug effects , Cell Adhesion Molecules/metabolism , Cells, Cultured , DNA-Binding Proteins/metabolism , Disease Models, Animal , Down-Regulation , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation/metabolism , Male , Mice , Monocytes/metabolism , Regulatory Factor X Transcription Factors , Stress, Mechanical , Transcription Factor CHOP/metabolism , Transcription Factors/metabolism , Ursodeoxycholic Acid/therapeutic use , X-Box Binding Protein 1
9.
Sci Rep ; 5: 12843, 2015 Aug 05.
Article in English | MEDLINE | ID: mdl-26243452

ABSTRACT

Extracorporeal shock wave (SW) therapy has been studied in the transfection of naked nucleic acids into various cell lines through the process of sonoporation, a process that affects the permeation of cell membranes, which can be an effect of cavitation. In this study, siRNAs were efficiently transfected into primary cultured cells and mouse tumor tissue via SW treatment. Furthermore SW-induced siRNA transfection was not mediated by SW-induced sonoporation, but by microparticles (MPs) secreted from the cells. Interestingly, the transfection effect of the siRNAs was transferable through the secreted MPs from human umbilical vein endothelial cell (HUVEC) culture medium after treatment with SW, into HUVECs in another culture plate without SW treatment. In this study, we suggest for the first time a mechanism of gene transfection induced by low-energy SW through secreted MPs, and show that it is an efficient physical gene transfection method in vitro and represents a safe therapeutic strategy for site-specific gene delivery in vivo.


Subject(s)
RNA, Small Interfering/genetics , Transfection/methods , Animals , Aorta , Cell Line, Tumor , Cell-Derived Microparticles/metabolism , Gene Knockdown Techniques , High-Energy Shock Waves , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mice, Inbred C57BL , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/prevention & control , Neovascularization, Physiologic , Primary Cell Culture , RNA Interference , Vascular Endothelial Growth Factor A/physiology
10.
Int J Cardiol ; 168(4): 4168-77, 2013 Oct 09.
Article in English | MEDLINE | ID: mdl-23915523

ABSTRACT

BACKGROUND: Extracorporeal shock wave has been used in the noninvasive treatment of various diseases including musculoskeletal disorders. In particular, shock wave with low energy level showed anti-inflammatory effect and increased angiogenesis in ischemic tissues. However, the detailed cellular pathway in endothelial signaling is not fully understood. We investigate the role of shock wave with low energy level in angiogenic gene expression and underlying molecular mechanism by comparing the laminar and oscillatory fluid shear stresses in endothelial cells. METHODS AND RESULTS: We show that shock wave with low energy level (0.012-0.045 mJ/mm(2)) stimulated phosphorylation of Akt, eNOS and Erk 1/2 in a time-dependent manner which is similar to the effect of laminar fluid shear stress. The transfection of endothelial cells with siRNA encoding VEGFR2, VE-cadherin and PECAM-1 inhibited shock wave-induced phosphorylation of Akt, eNOS and Erk 1/2 and angiogenic gene expressions, including Akt, eNOS, KLF2/4, and Nur77. Moreover, mechanical stimulation through extracorporeal shock wave induced endothelial cell migration and tube formation. CONCLUSIONS: Our results demonstrate that shock wave-induced Akt/eNOS phosphorylation and angiogenic gene expression were mediated through the mechanosensory complex formation involving VEGFR-2, VE-cadherin and PECAM-1 which was similar to the effect of laminar shear stress.


Subject(s)
Angiogenic Proteins/biosynthesis , Endothelial Cells/physiology , High-Energy Shock Waves , Mechanotransduction, Cellular/physiology , Membrane Fluidity/physiology , Shear Strength/physiology , Animals , Cell Movement/physiology , Cells, Cultured , Female , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/physiology , Humans , Male , Mice , Mice, Inbred C57BL
11.
Int J Mol Med ; 32(2): 373-80, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23708564

ABSTRACT

Soluble receptor for advanced glycation end products RAGE (sRAGE), a secretory form of RAGE, plays an important role in suppressing RAGE signals that induce pro-inflammatory gene activation in a range of inflammatory diseases, such as Alzheimer's disease, complications of diabetes mellitus and atherosclerosis. Recent studies have suggested that fluid shear stress generated by laminar blood flow protects blood vessels from atherosclerosis, whereas low and oscillatory shear stress (OSS) generated by disturbed blood flow causes atherosclerosis. Although RAGE levels are increased in atherosclerotic plaque, the regulatory mechanisms of sRAGE in the occurrence of atherosclerotic plaque induced by disturbed blood flow remain largely unknown. This study aimed to determine the effects of sRAGE as a competitive inhibitor of RAGE in atherogenesis induced by disturbed blood flow. To determine the role of sRAGE in atherosclerosis induced by disturbed blood flow, we used a mouse model of partial carotid artery ligation using ApoE(-/-) and C57BL/6 mice. Our results revealed that the expression of RAGE was significantly increased in the region of atherosclerotic plaque and that treatment with sRAGE attenuated the development of plaque formation. We found that the expression levels of RAGE and high mobility group box 1 (HMGB1), the agonistic ligand of RAGE, were significantly increased in human umbilical vein endothelial cells (HUVECs) under shear stress conditions induced by disturbed blood flow and suppressed following treatment with sRAGE. We further observed that treatment with sRAGE decreased the expression of vascular cell adhesion molecule­1 (VCAM-1) and markedly attenuated monocyte-endothelial cell adhesion. Taken together, our results reveal that sRAGE exerts anti-atherogenic effects by blocking the activation of the RAGE signaling pathway induced by disturbed blood flow and may thus be a potential therapeutic target for the prevention of atherosclerosis.


Subject(s)
Atherosclerosis/etiology , Atherosclerosis/metabolism , Hemodynamics , Mitogen-Activated Protein Kinases/metabolism , Animals , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Arteries/surgery , Cell Line , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Inflammation/etiology , Inflammation/metabolism , Male , Mice , Mice, Knockout , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
12.
J Nucl Med ; 53(11): 1779-85, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23038748

ABSTRACT

UNLABELLED: Radiolabeled lipophilic cationic compounds, such as (18)F-labeled phosphonium salt, accumulate in the mitochondria through a negative inner transmembrane potential. The purpose of this study was to develop and evaluate ((18)F-fluoropentyl)triphenylphosphonium salt ((18)F-FPTP) as a myocardial PET agent. METHODS: A reference compound of (18)F-FPTP was synthesized via 3-step nucleophilic substitution reactions and was radiolabeled via 2-step nucleophilic substitution reactions of no-carrier-added (18)F-fluoride. Accumulations of (18)F-FPTP, (3)H-tetraphenylphosphonium, and (99m)Tc-sestamibi were compared in a cultured embryonic cardiomyoblast cell line (H9c2). The biodistribution of (18)F-FPTP was assessed using BALB/c mice. The (18)F-FPTP small-animal PET study was performed in Sprague-Dawley rats with or without left coronary artery (LCA) ligation. RESULTS: (18)F-FPTP was synthesized with a radiochemical yield of 15%-20% and radiochemical purity of greater than 98%. Specific activity was greater than 6.3 TBq/µmol. Cell uptake of (18)F-FPTP was more than 15-fold higher in H9c2 than in normal fibroblasts (human normal foreskin fibroblasts). Selective collapse of mitochondrial membrane potential substantially decreased cellular uptake for (18)F-FPTP and (3)H-tetraphenylphosphonium, compared with that for (99m)Tc-sestamibi. The biodistribution data in mice (n = 24) showed rapid blood clearance and high accumulation in the heart. Heart-to-blood ratios at 10 and 30 min were 54 and 133, respectively. Heart-to-lung and heart-to-liver ratios at 10, 30, and 60 min were 4, 4, and 7 and 4, 5, and 7, respectively. Dynamic small-animal PET for 60 min after injection of (18)F-FPTP showed an initial spike of radioactivity, followed by retention in the myocardium and rapid clearance from the background. (18)F-FPTP small-animal PET images in LCA-occluded rats demonstrated sharply defined myocardial defects in the corresponding area of the myocardium. The myocardial defect size measured by (18)F-FPTP small-animal PET correlated closely with the hypoperfused area measured by quantitative 2,3,5-triphenyltetrazolium chloride staining (r(2) = 0.92, P < 0.001). CONCLUSION: The excellent pharmacokinetics of (18)F-FPTP and its correlation with 2,3,5-triphenyltetrazolium chloride staining in normal and LCA-occluded rats suggest that this molecular probe may have a high potential as a mitochondrial voltage sensor for PET. This probe may also allow high throughput, with multiple daily studies and a wide distribution of PET myocardial imaging in the clinic.


Subject(s)
Membrane Potential, Mitochondrial , Mitochondria/metabolism , Mitochondria/pathology , Myocardial Infarction/pathology , Organophosphorus Compounds/metabolism , Phosphines/metabolism , Animals , Biological Transport , Cell Line , Disease Models, Animal , Humans , Male , Mice , Mitochondria/diagnostic imaging , Myocardial Infarction/diagnostic imaging , Organophosphorus Compounds/chemical synthesis , Organophosphorus Compounds/pharmacokinetics , Phosphines/chemical synthesis , Phosphines/pharmacokinetics , Positron-Emission Tomography , Radiochemistry , Rats , Rats, Sprague-Dawley
13.
Blood ; 119(11): 2523-32, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22262759

ABSTRACT

The circulating peptide hormone hepcidin maintains systemic iron homeostasis. Hepcidin production increases during inflammation and as a result of endoplasmic reticulum (ER) stress. Elevated hepcidin levels decrease dietary iron absorption and promote iron sequestration in reticuloendothelial macrophages. Furthermore, increased plasma hepcidin levels cause hypoferremia and the anemia associated with chronic diseases. The signal transduction pathways that regulate hepcidin during inflammation and ER stress include the IL-6-dependent STAT-3 pathway and the unfolded protein response-associated cyclic AMP response element-binding protein-H (CREBH) pathway, respectively. We show that carbon monoxide (CO) suppresses hepcidin expression elicited by IL-6- and ER-stress agents by inhibiting STAT-3 phosphorylation and CREBH maturation, respectively. The inhibitory effect of CO on IL-6-inducible hepcidin expression is dependent on the suppressor of cytokine signaling-3 (SOCS-3) protein. Induction of ER stress in mice resulted in increased hepatic and serum hepcidin. CO administration inhibited ER-stress-induced hepcidin expression in vivo. Furthermore, ER stress caused iron accumulation in splenic macrophages, which could be prevented by CO. Our findings suggest novel anti-inflammatory therapeutic applications for CO, as well as therapeutic targets for the amelioration of anemia in the hypoferremic condition associated with chronic inflammatory and metabolic diseases.


Subject(s)
Antimetabolites/pharmacology , Antimicrobial Cationic Peptides/antagonists & inhibitors , Carbon Monoxide/pharmacology , Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , Endoplasmic Reticulum Stress/drug effects , Inflammation/pathology , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , Blotting, Western , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Hepcidins , Humans , Inflammation/drug therapy , Inflammation/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Iron/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Phosphorylation/drug effects , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction
14.
Mol Immunol ; 48(15-16): 1793-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21640381

ABSTRACT

The expression of C-reactive protein (CRP) rises rapidly in response to inflammation. The endoplasmic reticulum (ER) stress has been reported to cause CRP expression. Carbon monoxide (CO), a reaction product of heme oxygenase, exerts anti-inflammatory effects. In this study, we aimed to examine the role of CO in modulating ER stress-induced CRP expression. In HepG2 cells, ER stress triggered by tunicamycin, thapsigargin and homocysteine markedly induced CRP expression and the activation of protein kinase R-like endoplasmic reticulum kinase (PERK), inositol-requiring transmembrane kinase/endonuclease 1α (IRE1α), activating transcription factor 6 (ATF6), and hepatocyte-specific cyclic AMP response element binding protein H (CREBH). A CO-releasing molecule (CORM) inhibited ER stress-induced CRP expression. While CORM attenuated ER stress-induced activation of IRE1α, ATF6 and CREBH, it augmented PERK activation, which was associated with its inhibition of CRP expression. CORM also inhibited CRP expression in response to the pro-inflammatory cytokine IL-6 that was found to induce ER stress response in HepG2 cells. Moreover, in mice treated with the ER stress inducer tunicamycin, CORM administration reduced serum levels of CRP and the expression of CRP mRNA in the liver. Collectively, our findings suggest that CO may attenuate ER stress-induced CRP expression through modulation of the unfolded protein response.


Subject(s)
C-Reactive Protein/biosynthesis , Carbon Monoxide/metabolism , Endoplasmic Reticulum/metabolism , Inflammation/metabolism , Unfolded Protein Response/physiology , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Heme Oxygenase-1/metabolism , Hep G2 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Stress, Physiological/physiology , Transfection
15.
Blood Coagul Fibrinolysis ; 22(6): 532-40, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21670663

ABSTRACT

Combining thalidomide (Thal) with chemotherapeutic agents or steroid preparations led to improved response rates in the treatment of multiple myeloma. However, deep vein thrombosis (DVT) is one of the most serious side-effects noted with this regimen, and how a Thal-based regimen causes DVT is unclear. We investigated the procoagulant effects of Thal when combined with chemotherapeutic agents in vitro, focusing on tissue factor (TF) and phosphatidylserine. We examined the effects of the chemotherapeutic doxorubicin hydrochloride (Dox) and the steroid dexamethasone (Dex), with or without Thal. Our study used the human vascular endothelial, monocytic, and myeloma cell lines, EAhy926, THP-1, and RPMI8226, respectively. In EAhy926 and THP-1, Dex treatment increased expression of TF, which may induce procoagulant activity (PCA). Upregulation of TF mRNA correlated with activation of the Egr-1 pathway. In Thal and Dex treatments, the increase of PCA induction from phosphatidylserine exposure was modest. In contrast, Dox and Thal-Dox increased phosphatidylserine exposure in both cell types. In THP-1 cells, cell surface phosphatidylserine exposure correlated with increased PCA by Dox. Thal alone showed a modest increase in phosphatidylserine exposure in endothelial cells and monocytes. When Thal is given in combination with chemotherapies or Dex, endothelial cell and monocyte PCA may be induced through phosphatidylserine exposure, or TF expression. Induction may be protracted by Thal, which has an antiangiogenic activity. Therefore, prophylactic anticoagulant strategies should be considered in Thal-based combination regimens.


Subject(s)
Antineoplastic Agents/pharmacology , Blood Coagulation/drug effects , Dexamethasone/adverse effects , Doxorubicin/adverse effects , Phosphatidylserines/analysis , Thalidomide/adverse effects , Thromboplastin/biosynthesis , Annexin A5/analysis , Anticoagulants/pharmacology , Antineoplastic Agents/adverse effects , Blood Coagulation Factors/analysis , Blood Coagulation Factors/metabolism , Cell Line , Dexamethasone/pharmacology , Doxorubicin/pharmacology , Drug Combinations , Drug Interactions , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Flow Cytometry , Humans , Monocytes/drug effects , Monocytes/metabolism , Multiple Myeloma/complications , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Thalidomide/pharmacology , Thromboplastin/analysis , Venous Thrombosis/chemically induced , Venous Thrombosis/complications
17.
Thromb Haemost ; 98(2): 368-74, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17721619

ABSTRACT

The extrinsic coagulation system initiated by tissue factor (TF) appears to be a major procoagulant stimulus during cardiopulmonary bypass (CPB), although the precise mechanisms remain to be revealed. We recently reported the appearance of TF-bearing leukocytes during CPB and described their role in promoting coagulation. In this study, we visually identified the in-vivo appearance of TF-bearing leukocytes and platelet-derived particles on leukocytes in the pericardial blood during cardiac surgery with CPB, by flow cytometry and immunoelectron microscopy. Preliminary flow cytometric experiments showed that the proportion of TF-positive or both TF- and platelet antigen CD41a-positive leukocytes was increased markedly in pericardial blood obtained during CPB, compared with the proportions in preoperative circulating blood. Immunoelectron microscopic analysis revealed that both monocytes and polymorphonuclear leukocytes in the pericardial blood express TF. On the surfaces of these cells, CD41a-positive or both CD41a- and TF-positive platelet-derived particles were observed. Platelet-derived particles include not only microparticles, but also platelets themselves. Leukocytes from preoperative circulating blood contained far fewer of these particles. Our results demonstrate the in-vivo appearance of TF-bearing platelet-derived particles on leukocytes during cardiac surgery with CPB. These findings may be important for the development of strategies to control procoagulant activities during and after cardiac surgery.


Subject(s)
Blood Platelets/metabolism , Cardiopulmonary Bypass , Leukocytes/metabolism , Membrane Microdomains/metabolism , Platelet Membrane Glycoprotein IIb/analysis , Thromboplastin/analysis , Blood Platelets/chemistry , Blood Platelets/ultrastructure , Flow Cytometry , Humans , Leukocytes/chemistry , Microscopy, Immunoelectron , Monocytes/chemistry , Neutrophils/chemistry , Particle Size
18.
Lab Invest ; 87(6): 540-7, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17401435

ABSTRACT

An essential coagulation factor, tissue factor (TF), is rapidly expressed by human monocytes when exposed to a variety of agonists, such as lipopolysaccharide or tumor necrosis factor (TNF). We previously found that 1alpha,25-dihydroxyvitamin D(3) (1,25(OH)(2)D(3)) and its potent synthetic analogs downregulate TF and upregulate thrombomodulin expression on monocytic cells, counteracting the effects of TNF at the level of transcription. The human TF gene has characteristic binding sequences for activator protein-1 (AP-1) (c-Jun/c-Fos), nuclear factor-kappaB (NF-kappaB), Sp-1, and early growth response factor-1 (Egr-1). In this study, we investigated the regulatory mechanisms by which 1,25(OH)(2)D(3) inhibits TNF-induced TF expression in human monocytic cells. 1,25(OH)(2)D(3) reduced basal and TNF-induced TF activities. Gel-shift assay and luciferase assay with the respective reporter vectors showed that 1,25(OH)(2)D(3) reduced basal and TNF-induced activities of the nuclear proteins AP-1 and NF-kappaB, but not Egr-1. 1,25(OH)(2)D(3) inhibited TNF-induced phosphorylation of c-Jun without affecting phosphorylation of the other pathways. On the other hand, 1,25(OH)(2)D(3) directly inhibited nuclear binding and activities of NF-kappaB in the nucleus without affecting phosphorylation of the NF-kappaB activation pathway. These results indicate that 1,25(OH)(2)D(3) suppresses basal and TNF-induced TF expression in monocytic cells by inhibition of AP-1 and NF-kappaB activation pathways, but not of Egr-1. Our results may help to elucidate the regulatory mechanisms of 1,25(OH)(2)D(3) in TF induction, and may have physiological significance in the clinical challenge to use potential 1,25(OH)(2)D(3) analogs in antithrombotic therapy as well as immunomodulation and antineoplastic therapy of leukemia.


Subject(s)
Dihydrotachysterol/analogs & derivatives , Monocytes/metabolism , NF-kappa B/antagonists & inhibitors , Thromboplastin/antagonists & inhibitors , Transcription Factor AP-1/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology , Cells, Cultured , Dihydrotachysterol/pharmacology , Gene Expression Regulation/drug effects , Humans , Monocytes/drug effects , Thromboplastin/genetics
19.
Med Sci Monit ; 12(2): BR51-6, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16449942

ABSTRACT

BACKGROUND: Ischemia-reperfusion injury of the lungs seems to be initiated by the activation of alveolar macrophages, and mediated by matrix metalloproteinases (MMP)s, although their roles have not been fully elucidated. Therefore, we used a novel MMP inhibitor (ONO-4817) with high affinities for MMP-2 and MMP-9 to investigate the roles of MMPs in reperfusion injury of the lungs. MATERIAL/METHODS: After 18 h of cold ischemia, isolated rat lungs were ventilated and reperfused. Lungs without ischemia served as controls. Lungs were reperfused with fresh blood with or without the MMP inhibitor for 120 min at 37 degrees C. RESULTS: The oxygenation capacity of lungs after ischemia deteriorated progressively during 120 min of reperfusion, but was preserved by the MMP inhibitor (p<0.05). The histopathology of the lungs after ischemia-reperfusion showed interstitial edema accompanied by neutrophil migration, and the number of neutrophils, but not macrophages, in the broncho-alveolar lavage increased to more than two-fold the value in control lungs without ischemia (p<0.01). These changes were attenuated by the MMP inhibitor (p<0.01). Similarly, an increase in the tissue malondialdehyde level in lungs after ischemia-reperfusion was ameliorated by the MMP inhibitor (p<0.01). The expressions of CD11c and CD31 adhesion molecules in extracted alveolar macrophages increased in lungs after ischemia-reperfusion compared with control lungs without ischemia, and the MMP inhibitor had no obvious effect. CONCLUSIONS: Our data show that ONO-4817 prevented neutrophil migration into the interstitial space and alveolus in the lungs, and reduced the production of oxygen free radicals, suggesting that this is an important mechanism in reperfusion injury.


Subject(s)
Lung Injury , Lung/drug effects , Matrix Metalloproteinase Inhibitors , Phenyl Ethers/pharmacology , Protease Inhibitors/pharmacology , Reperfusion Injury/enzymology , Reperfusion Injury/prevention & control , Animals , CD11c Antigen/metabolism , In Vitro Techniques , Lung/pathology , Lung/physiopathology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/pathology , Male , Neutrophils/pathology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology
20.
Eur J Cardiothorac Surg ; 26(5): 995-1001, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15519195

ABSTRACT

OBJECTIVE: Immediate loss of thrombomodulin activity in the endothelium of vein grafts has been demonstrated during 90 min exposure to arterial circulation; this loss of activity is ascribed as an important cause of early thrombosis. Conventional ex vivo gene transfection after vein harvest cannot cover this acute period immediately after implantation. We have established a highly efficient non-viral gene therapy protocol utilizing modified transferrin receptor-facilitated gene transfer. Using this technique, we examined whether in vivo thrombomodulin gene therapy, directed to the endothelium of rat veins 2 days prior to grafting, may prevent thromboresistance impairment of vein grafts under simulated arterial circulation. METHODS: Abdomen of SD rat was opened and cationic liposome:transferrin:thrombomodulin gene complexes or the vector without DNAs were applied to the inferior vena cava of rats while blood flow was reduced by proximal and distal clamping. After 2 days, the transfected veins were harvested and thrombomodulin expression and thromboresistance properties determined before and after exposure to an artificial circuit. RESULTS: The trial of gene transfection using variable doses of DNAs confirmed that 7.5 microg of total DNAs was the most efficient quantity for thrombomodulin gene transfection to IVCs, although accompanying an increase of gene expression in other downstream organs. By transfection of the thrombomodulin gene in IVCs, the generation capacity of activated protein C in venous endothelium increased three-fold compared with veins treated with vector alone (P<0.01). Under simulated arterial circulation, perfusion of veins treated with vector alone decreased thrombomodulin activity to 36% of preperfused levels (P<0.01), whereas transfected grafts preserved the activity at normal vein endothelium levels even after perfusion. Consequently, the increase in endothelial thrombin activity induced by simulated arterial circulation was markedly attenuated in transfected veins (P<0.01), while immunohistochemistry confirmed the preservation of endothelial lining. CONCLUSIONS: Transferrin receptor-facilitated in vivo gene transfer to the inferior vena cava resulted in sufficient thrombomodulin gene expression immediately after graft implantation and subsequent maintenance of thromboresistance even after exposure to arterial pressure. Although further studies are needed, the present results suggest the possibility of gene therapy targeting acute phases of vein graft disease.


Subject(s)
Gene Transfer Techniques , Genetic Therapy/methods , Graft Occlusion, Vascular/prevention & control , Thrombomodulin/metabolism , Animals , Blotting, Western , Endothelium, Vascular/metabolism , Graft Occlusion, Vascular/metabolism , Liposomes , Male , Rats , Rats, Sprague-Dawley , Receptors, Transferrin/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Thrombomodulin/genetics , Vena Cava, Inferior/metabolism , Vena Cava, Inferior/transplantation , Venous Thrombosis/metabolism , Venous Thrombosis/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL
...