Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Korean J Physiol Pharmacol ; 19(6): 549-55, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26557022

ABSTRACT

We attempted to investigate molecular mechanisms underlying phenotypic change of vascular smooth muscle cells (VSMCs) by determining signaling molecules involved in chemokine production. Treatment of human aortic smooth muscle cells (HAoSMCs) with thrombin resulted not only in elevated transcription of the (C-C motif) ligand 11 (CCL11) gene but also in enhanced secretion of CCL11 protein. Co-treatment of HAoSMCs with GF109230X, an inhibitor of protein kinase C, or GW5074, an inhibitor of Raf-1 kinase, caused inhibition of ERK1/2 phosphorylation and significantly attenuated expression of CCL11 at transcriptional and protein levels induced by thrombin. Both Akt phosphorylation and CCL11 expression induced by thrombin were attenuated in the presence of pertussis toxin (PTX), an inhibitor of Gi protein-coupled receptor, or LY294002, a PI3K inhibitor. In addition, thrombin-induced production of CCL11 was significantly attenuated by pharmacological inhibition of Akt or MEK which phosphorylates ERK1/2. These results indicate that thrombin is likely to promote expression of CCL11 via PKC/Raf-1/ERK1/2 and PTX-sensitive protease-activated receptors/PI3K/Akt pathways in HAoSMCs. We propose that multiple signaling pathways are involved in change of VSMCs to a secretory phenotype.

2.
J Neurosci Res ; 92(2): 206-17, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24254769

ABSTRACT

ß-Amyloid (Aß) deposits and hyperphosphorylated tau aggregates are the chief hallmarks in the Alzheimer's disease (AD) brains, but the strategies for controlling these pathological events remain elusive. We hypothesized that CK2-coupled SIRT1 activation stimulated by cilostazol suppresses tau acetylation (Ac-tau) and tau phosphorylation (P-tau) by inhibiting activation of P300 and GSK3ß. Aß was endogenously overproduced in N2a cells expressing human APP Swedish mutation (N2aSwe) by exposure to medium containing 1% fetal bovine serum for 24 hr. Increased Aß accumulation was accompanied by increased Ac-tau and P-tau levels. Concomitantly, these cells showed increased P300 and GSK3ß P-Tyr216 expression; their expressions were significantly reduced by treatment with cilostazol (3-30 µM) and resveratrol (20 µM). Moreover, decreased expression of SIRT1 and its activity by Aß were significantly reversed by cilostazol as by resveratrol. In addition, cilostazol strongly stimulated CK2α phosphorylation and its activity, and then stimulated SIRT1 phosphorylation. These effects were confirmed by using the pharmacological inhibitors KT5720 (1 µM, PKA inhibitor), TBCA (20 µM, inhibitor of CK2), and sirtinol (20 µM, SIRT1 inhibitor) as well as by SIRT1 gene silencing and overexpression techniques. In conclusion, increased cAMP-dependent protein kinase-linked CK2/SIRT1 expression by cilostazol can be a therapeutic strategy to suppress the tau-related neurodegeneration in the AD brain.


Subject(s)
Keratin-2/metabolism , Neurons/drug effects , Neuroprotective Agents/pharmacology , Sirtuin 1/biosynthesis , Tauopathies/metabolism , Tetrazoles/pharmacology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Blotting, Western , Cell Line , Cilostazol , Fluorescent Antibody Technique , Humans , Mice , Neurons/metabolism , Transfection , tau Proteins/metabolism
3.
J Neurosci Res ; 92(11): 1581-90, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24903973

ABSTRACT

The accumulation of plaques of ß-amyloid (Aß) peptides, a hallmark of Alzheimer's disease, results from the sequential cleavage of amyloid precursor protein (APP) by activation of ß- and γ-secretases. However, the production of Aß can be avoided by alternate cleavage of APP by α-and γ-secretases. We hypothesized that cilostazol attenuates Aß production by increasing a disintegrin and metalloproteinase 10 (ADAM10)/α-secretase activity via SIRT1-coupled retinoic acid receptor-ß (RARß) activation in N2a cells expressing human APP Swedish mutation (N2aSwe). To evoke endogenous Aß overproduction, the culture medium was switched from medium containing 10% fetal bovine serum (FBS) to medium containing 1% FBS, and cells were cultured for 3∼24 hr. After depletion of FBS in media, N2aSwe cells showed increased accumulations of full-length APP (FL-APP) and Aß in a time-dependent manner (3-24 hr) in association with decreased ADAM10 protein expression. When pretreated with cilostazol (10-30 µM), FL-APP and Aß levels were significantly reduced, and ADAM10 and α-secretase activities were restored. Furthermore, the effect of cilostazol on ADAM10 expression was antagonized by pretreating Rp-cAMPS and sirtinol and by SIRT1-gene silencing. In the N2aSwe cells overexpressing the SIRT1 gene, ADAM10, and sAPPα levels were significantly elevated. In addition, like all-trans retinoic acid, cilostazol enhanced the protein expressions of RARß and ADAM10, and the cilostazol-stimulated ADAM10 elevation was significantly attenuated by LE135 (a RARß inhibitor), sirtinol, and RARß-gene silencing. In conclusion, cilostazol suppresses the accumulations of FL-APP and Aß by activating ADAM10 via the upregulation of SIRT1-coupled RARß.


Subject(s)
Amyloid beta-Peptides/metabolism , Matrix Metalloproteinase 10/metabolism , Neuroprotective Agents/pharmacology , Peptide Fragments/metabolism , Receptors, Retinoic Acid/metabolism , Sirtuin 1/metabolism , Tetrazoles/pharmacology , Up-Regulation/drug effects , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Benzamides/pharmacology , Cell Line, Tumor , Cilostazol , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Disintegrins , Enzyme Inhibitors/pharmacology , Humans , Mice , Naphthols/pharmacology , Neuroblastoma/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sirtuin 1/genetics , Thionucleotides/pharmacology , Time Factors , Transfection
4.
Korean J Physiol Pharmacol ; 18(6): 475-80, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25598661

ABSTRACT

We investigated the question of whether cholesterol catabolite can influence expression of inflammatory cytokines via Toll-like receptors (TLR) in monocytic cells. Treatment of THP-1 monocytic cells with 27-hydroxycholesterol (27OHChol) resulted in induction of gene transcription of TLR6 and elevated level of cell surface TLR6. Addition of FSL-1, a TLR6 agonist, to 27OHChol-treated cells resulted in transcription of the IL-1α gene and enhanced secretion of the corresponding gene product. However, cholesterol did not affect TLR6 expression, and addition of FSL-1 to cholesterol-treated cells did not induce expression of IL-1α. Using pharmacological inhibitors, we investigated molecular mechanisms underlying the expression of TLR6 and IL-1α. Treatment with Akt inhibitor IV or U0126 resulted in significantly attenuated expression of TLR6 and IL-1α induced by 27OHChol and 27OHChol plus FSL-1, respectively. In addition, treatment with LY294002, SB202190, or SP600125 resulted in significantly attenuated secretion of IL-1α. These results indicate that 27OHChol can induce inflammation by augmentation of TLR6-mediated production of IL-1α in monocytic cells via multiple signaling pathways.

5.
Biochem Biophys Res Commun ; 437(3): 392-6, 2013 Aug 02.
Article in English | MEDLINE | ID: mdl-23831464

ABSTRACT

The polyene antifungal antibiotic nystatin can interact with cholesterol, thereby altering the composition of the plasma membrane in eukaryotic cells. We investigated whether nystatin influences responses to the infection by inducing expression of chemokines. THP-1 macrophages rarely expressed CC chemokine ligand 2 (CCL2) and CXCL8. However, nystatin dose-dependently increased CCL2 and CXCL8 expression at the mRNA and protein levels. To understand the molecular mechanisms of the antifungal agent, we identified cellular factors activated by nystatin and those involved in nystatin-induced upregulation of CCL2 and CXCL8. Treatment with nystatin resulted in enhanced phosphorylation of Akt, ERK1/2, p38 MAPK, and JNK. Treatment with cholesterol, LY294002, Akt inhibitor IV, U0126, and SP6001250 resulted in abrogation or significant attenuation of nystatin-induced CCL2 expression. Nystatin-mediated CXCL8 expression was attenuated in the presence of Akt inhibitor IV and SP6001250. These results indicate that exposure of human macrophages to nystatin can lead to differential regulation of CCL2 and CXCL8 via the activation of multiple cellular kinases. We propose that upregulation of CCL2 and CXCL8 contributes to pharmacological effects of nystatin.


Subject(s)
Antifungal Agents/pharmacology , Chemokine CCL2/biosynthesis , Interleukin-8/biosynthesis , Macrophages/drug effects , Macrophages/metabolism , Nystatin/pharmacology , Cell Line , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Humans , Inflammation Mediators/metabolism , Inflammation Mediators/pharmacology , Interleukin-8/genetics , Interleukin-8/metabolism , Macrophages/microbiology , Up-Regulation/drug effects , Up-Regulation/genetics
6.
J Neurosci Res ; 90(8): 1566-76, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22422579

ABSTRACT

Amyloid-ß peptide (Aß) deposits in the brain are critical in the neurotoxicity induced by Aß. This study elucidates the underlying signaling pathway by which cilostazol protects HT22 neuronal cells from Aß(1-40) (3-30 µM)-induced deterioration of cell proliferation, viability, and neurite elongation. Cilostazol rescued HT22 cells from the apoptotic cell death induced by Aß toxicity through the downregulation of phosphorylated p53 (Ser15), Bax, and caspase-3 and the upregulation of Bcl-2 expression, which improved neuronal cell proliferation and viability. Furthermore, Aß(1-40) suppressed both phosphorylated CK2α protein expression and CK2 activity in the cytosol; these were concentration dependently recovered by cilostazol (3-30 µM). Cilostazol significantly increased the levels of GSK-3ß phosphorylation at Ser9 and ß-catenin phosphorylation at Ser675 in the cytosol and nucleus. Cilostazol effects were reversed by KT5720 (1 µM, PKA inhibitor) and TBCA (40 µM, inhibitor of CK2) and CK2α knockdown by siRNA transfection. Likewise, Aß-stimulated GSK-3ß phosphorylation at Tyr 216 was decreased by cilostazol in the control but not in the CK2α siRNA-transfected cells. Furthermore, the Aß (10 µM)-induced suppression of neurite elongation was recovered by cilostazol; this recovery was attenuated by inhibitors such as KT5720 and TBCA and blocked by CK2α knockdown. In conclusion, increased cAMP-dependent protein kinase-linked CK2α activation underlies the pharmacological effects of cilostazol in downregulating p53 phosphorylation at Ser15 and upregulating GSK-3ß phosphorylation at Ser9/ß-catenin phosphorylation at Ser675, thereby suppressing Aß(1-40)-induced neurotoxicity and improving neurite elongation.


Subject(s)
Amyloid beta-Peptides/toxicity , Casein Kinase II/metabolism , Neurites/drug effects , Neuroprotective Agents/pharmacology , Tetrazoles/pharmacology , Animals , Apoptosis , Blotting, Western , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Cilostazol , Enzyme Activation/drug effects , Gene Knockdown Techniques , Hippocampus/drug effects , Hippocampus/metabolism , Mice , Neurites/metabolism , RNA, Small Interfering , Signal Transduction/drug effects , Transfection
7.
J Cardiovasc Pharmacol ; 60(2): 199-207, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22622234

ABSTRACT

Atherosclerotic plaque contains materials, such as cholesterol, oxysterols, cell debris, modified fatty acids, and infiltrated cells. Among them, cholesterol is the major component in plaque. Cholesterol is known to originate from the influx of extracellular materials, but this explanation is not enough for the cholesterol accumulation observed in atherosclerotic plaque. This study examined the origins of cholesterols in plaques. The main focus was to determine if the intracellular cholesterol levels are affected by oxysterols in human vascular smooth muscle cells. The results showed that the cholesterol levels increased in response to a 7-ketocholesterol (7K)-treatment in a dose-dependent manner. Eight enzymes involved in cholesterol biosynthesis were examined. Among them, squalene epoxidase (SQLE) was increased by 7K but not by 7α-hydroxycholesterol, 27-hydroxycholesterol (27OH-chol), or cholesterol. The 7K-induced SQLE expression was suppressed in the presence of the enzyme inhibitor SB203580 but not by UO126 and SP600125. The SQLE immunoreactivity was detected in the atherosclerotic plaque of the aortic roots from apoE mice. In addition, 7K increased the cholesterol level and SQLE expression in murine bone marrow-derived macrophages. This suggests that 7K increases the intracellular cholesterol level through an elevation of SQLE expression, which might affect the progress of cholesterol accumulation in the atherosclerotic lipid core.


Subject(s)
Aortic Diseases/metabolism , Atherosclerosis/metabolism , Cholesterol/metabolism , Ketocholesterols/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Cells, Cultured , Cholesterol/biosynthesis , Disease Models, Animal , Homeostasis , Humans , Imidazoles/pharmacology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Plaque, Atherosclerotic , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Squalene Monooxygenase/metabolism , Time Factors , Up-Regulation
8.
Korean J Physiol Pharmacol ; 16(6): 447-53, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23271927

ABSTRACT

TLR6 forms a heterodimer with TLR2 and TLR4. While proinflammatory roles of TLR2 and TLR4 are well documented, the role of TLR6 in inflammation is poorly understood. In order to understand mechanisms of action of TLR6 in inflammatory responses, we investigated the effects of FSL-1, the TLR6 ligand, on expression of chemokine CCL2 and cytokine IL-1ß and determined cellular factors involved in FSL-1-mediated expression of CCL2 and IL-1ß in mononuclear cells. Exposure of human monocytic leukemia THP-1 cells to FSL-1 resulted not only in enhanced secretion of CCL2 and IL-1ß, but also profound induction of their gene transcripts. Expression of CCL2 was abrogated by treatment with OxPAPC, a TLR-2/4 inhibitor, while treatment with OxPAPC resulted in partially inhibited expression of IL-1ß. Treatment with FSL-1 resulted in enhanced phosphorylation of Akt and mitogen-activated protein kinases and activation of protein kinase C. Treatment with pharmacological inhibitors, including SB202190, SP6001250, U0126, Akt inhibitor IV, LY294002, GF109203X, and RO318220 resulted in significantly attenuated FSL-1-mediated upregulation of CCL2 and IL-1ß. Our results indicate that activation of TLR6 will trigger inflammatory responses by upregulating expression of CCL2 and IL-1ß via TLR-2/4, protein kinase C, PI3K-Akt, and mitogen-activated protein kinases.

9.
Biochem Biophys Res Commun ; 408(1): 132-8, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21463602

ABSTRACT

Peptidoglycan (PG) is detected in a high proportion in inflammatory cell-rich regions of human atheromatous plaques. In the present study, we determined the cellular factors involved in PG-mediated chemokine expression in mononuclear cells in order to understand the molecular mechanisms of inflammatory responses to bacterial pathogen-associated molecular patterns in the diseased artery. Exposure of human monocytic leukemia THP-1 cells to PG resulted in not only enhanced secretion of CCL2 and CCL4 but also profound induction of their gene transcripts, which were abrogated by oxidized 1-palmitoyl-2-arachidonosyl-sn-phosphatidylcholine, an inhibitor of Toll-like receptors (TLRs)-2/4, but not by polymyxin B. PG enhanced phosphorylation of Akt and mitogen-activated protein kinases and activated protein kinase C. Pharmacological inhibitors such as SB202190, SP6001250, U0126, Akt inhibitor IV, rapamycin, and RO318220 significantly attenuated PG-mediated up-regulation of CCL2 and CCL4. We propose that PG contributes to vascular inflammation in atherosclerotic plaques by upregulating expression of mononuclear cell chemoattractants via TLR-2, protein kinase C, Akt, mTOR, and mitogen-activated protein kinases.


Subject(s)
Chemokine CCL2/metabolism , Chemokine CCL4/metabolism , Chemotactic Factors/metabolism , Monocytes/immunology , Peptidoglycan/immunology , Cell Line, Tumor , Chemokine CCL2/genetics , Chemokine CCL4/genetics , Humans , Monocytes/drug effects , Peptidoglycan/pharmacology , Phospholipid Ethers/pharmacology , Signal Transduction , Toll-Like Receptor 2/antagonists & inhibitors , Toll-Like Receptor 4/antagonists & inhibitors , Transcription, Genetic
10.
J Neurosci Res ; 89(5): 650-60, 2011 May.
Article in English | MEDLINE | ID: mdl-21337364

ABSTRACT

Recruitment and adhesion of exogenous endothelial progenitor cells (EPCs) or endogenously mobilized bone marrow mononuclear cells (BM MNCs) to the sites of ischemia is an important focus of cell therapy. This study sought to determine whether cilostazol enhances integrin-dependent homing of progenitor cells both in vitro and in vivo. In the in vitro experiments with human umbilical cord blood (HUCB)-derived EPCs, cilostazol (10 µM) stimulated up-regulation of integrins ß1, α1, and αv as well as 8-pCPT-2'-O-Me-cAMP (100 µM; 8-pCPT, Epac activator). Cilostazol and 8-pCPT significantly enhanced migration and adhesion of HUCB EPCs to a fibronectin-coated plate and endothelial cells, which were inhibited by KT5720 (PKA inhibitor, 1 µM) and GGTI-298 (Rap1 inhibitor, 20 µM). Cilostazol stimulated Epac1 expression and up-regulated the active Rap1, as did 8-pCPT, and they were suppressed by KT5720 (P < 0.001) and GGTI-298 (P < 0.001). 8-pCPT increased p-CREB expression and stimulated PKA activity, which was inhibited by KT5720, Rp-cAMPS, and GGTI-298. In addition, N(6)-benzoyl-cAMP (100 µM) increased Rap1 GTP expression, as did 8-pCPT; they were suppressed by Rp-cAMPS and GGTI-298. The in vivo experiments showed that cilostazol (30 mg/kg/day, orally for 7 days) significantly enhanced the integrin ß1 expression in the molecular layer and up-regulated homing of BM MNCs to the injured molecular layer with increased capillary density in mouse brain subjected to transient forebrain ischemia (n = 6, P < 0.001). In conclusion, cilostazol stimulated integrin expression and enhanced migration and adhesion of progenitor cells through cooperative activation of PKA and Epac signals; such activity may improve the efficacy of cell therapy for ischemic disease.


Subject(s)
Chemotaxis/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Integrin beta1/physiology , Mesenchymal Stem Cells/drug effects , Tetrazoles/pharmacology , Animals , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain Ischemia/therapy , Cells, Cultured , Chemotaxis/physiology , Cilostazol , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Disease Models, Animal , Humans , Infant, Newborn , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Protein Kinase Inhibitors/pharmacology , Tetrazoles/therapeutic use , Up-Regulation/drug effects , Up-Regulation/physiology
11.
Arthritis Rheum ; 62(3): 732-41, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20131233

ABSTRACT

OBJECTIVE: To assess the effects of cilostazol in inhibiting proliferation and enhancing apoptosis in synovial cells from patients with rheumatoid arthritis (RA). METHODS: Synovial cell proliferation was measured by MTT assay. The expression of NF-kappaB, IkappaBalpha, Bcl-2, Bax, heme oxygenase 1 (HO-1), and Nrf2 was determined by Western blotting. RESULTS: Cilostazol suppressed synovial cell proliferation by arresting the G(2)/M phases of the cell cycle, and this was reversed by KT5720, an inhibitor of protein kinase A. Cilostazol increased the number of TUNEL-positive cells, with increased cytochrome c release and apoptosis-inducing factor translocation as well as increased caspase 3 activation. Cilostazol (10 microM) and cobalt protoporphyrin IX (CoPP) increased HO-1 messenger RNA and protein expression. These effects were suppressed by zinc protoporphyrin IX (ZnPP), an HO-1 inhibitor. Cilostazol and CoPP significantly increased IkappaBalpha in the cytosol and decreased NF-kappaB p65 expression in the nucleus. Increased expression of tumor necrosis factor alpha (TNFalpha), interleukin-1beta (IL-1beta), and IL-6 induced by lipopolysaccharide was attenuated by cilostazol and CoPP, and this was reversed by ZnPP. In mice with collagen-induced arthritis treated with cilostazol (10 and 30 mg/kg/day), paw thickness was decreased with increased apoptotic cells in the joints. In synovial cells transfected with small interfering RNA (siRNA) targeting HO-1, cilostazol did not suppress expression of TNFalpha, IL-1beta, and IL-6, in contrast to findings with negative control cells. Cilostazol- and CoPP-induced HO-1 expression was diminished in cells transfected with Nrf2 siRNA. CONCLUSION: Cilostazol suppressed proliferation of synovial cells from RA patients by enhancing apoptosis, and also inhibited cytokine production via mediation of cAMP-dependent protein kinase activation-coupled Nrf2-linked HO-1 expression.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cytokines/biosynthesis , Heme Oxygenase-1/biosynthesis , NF-E2-Related Factor 2/metabolism , Phosphodiesterase Inhibitors/pharmacology , Synovial Membrane/drug effects , Tetrazoles/pharmacology , Animals , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Blotting, Western , Cell Division/drug effects , Cilostazol , Enzyme Induction , Mice , Mice, Inbred DBA , Synovial Membrane/cytology
12.
J Neurosci Res ; 88(10): 2228-38, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20175201

ABSTRACT

Cilostazol is known to be a specific type III phosphodiesterase inhibitor, which promotes increased intracellular cAMP levels. We assessed the effect of cilostazol on production of angioneurins and chemokines and recruitment of new endothelial cells for vasculogenesis in a mouse model of transient forebrain ischemia. Pyramidal cell loss was prominently evident 3-28 days postischemia, which was markedly ameliorated by cilostazol treatment. Expression of angioneurins, including endothelial nitric oxide synthase, vascular endothelial growth factor, and brain-derived neurotrophic factor, was up-regulated by cilostazol treatment in the postischemic hippocampus. Cilostazol also increased Sca-1/vascular endothelial growth factor receptor-2 positive cells in the bone marrow and circulating peripheral blood and the number of stromal cell-derived factor-1alpha-positive cells in the molecular layer of the hippocampus, which colocalized with CD31. CXCR4 chemokine receptors were up-regulated by cilostazol in mouse bone marrow-derived endothelial progenitor cells, suggesting that cilostazol may be important in targeting or homing in of bone marrow-derived stem cells to areas of injured tissues. CD31-positive cells were colocalized with almost all bromodeoxyuridine-positive cells in the molecular layer, indicating stimulation of endothelial cell proliferation by cilostazol. These data suggest that cilostazol markedly enhances neovascularization in the hippocampus CA1 area in a mouse model of transient forebrain ischemia, providing a beneficial interface in which both bone marrow-derived endothelial progenitor cells and angioneurins influence neurogenesis in injured tissue. (c) 2010 Wiley-Liss, Inc.


Subject(s)
Hippocampus/drug effects , Ischemic Attack, Transient/drug therapy , Neovascularization, Physiologic/drug effects , Phosphodiesterase Inhibitors/pharmacology , Tetrazoles/pharmacology , Adult Stem Cells/drug effects , Adult Stem Cells/physiology , Animals , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/pathology , CA1 Region, Hippocampal/physiopathology , Cell Death/drug effects , Cell Proliferation/drug effects , Cilostazol , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/physiology , Hippocampus/pathology , Hippocampus/physiopathology , Ischemic Attack, Transient/pathology , Ischemic Attack, Transient/physiopathology , Male , Mice , Mice, Inbred C57BL , Prosencephalon/drug effects , Prosencephalon/pathology , Prosencephalon/physiopathology
13.
Biochem Biophys Res Commun ; 378(3): 444-9, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-19028451

ABSTRACT

Oxidative stress results in sustained release of heat shock protein 90 (HSP90) from vascular smooth muscle cells (VSMCs). The aim of this article is to investigate whether extracellular HSP90 predisposes VSMCs to pro-inflammatory phenotype. Exposure of aortic smooth muscle cells to HSP90 elevated IL-8 release and IL-8 transcript via promoter activation. HSP90-induced IL-8 promoter activation was suppressed by dominant-negative forms of Toll-like receptor (TLR)-4 and MyD88, but not by dominant-negative-forms of TLR-3, TLR-2, and TRIF. IL-8 up-regulation in response to HSP90 was also attenuated by IkappaB, rasveratrol, curcumin, diphenyleneiodium, N-acetylcystein, U0126, and SB202190. Mutation at the NF-kappaB- or C/EBP-binding site, but not at the AP-1-binding site, in the IL-8 promoter region suppressed the promoter activation by HSP90. This study proposes that extracellular HSP90 would contribute to IL-8 elevation in the stressed vasculature, and that TLR-4, mitogen-activated protein kinases, NF-kappaB, and reactive oxygen species are involved in that process.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Interleukin-8/genetics , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Transcriptional Activation , CCAAT-Enhancer-Binding Proteins/metabolism , HSP90 Heat-Shock Proteins/pharmacology , Humans , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , NF-kappa B/metabolism , Promoter Regions, Genetic/drug effects , Reactive Oxygen Species/metabolism , Toll-Like Receptor 4/metabolism , Transcription, Genetic
14.
J Pharmacol Exp Ther ; 329(2): 571-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19221061

ABSTRACT

In a previous study, cilostazol promoted differentiation of 3T3-L1 fibroblasts into adipocytes and improved insulin sensitivity by stimulating peroxisome proliferator-activated receptor (PPAR) gamma transcription. This study evaluated the in vivo efficacy of cilostazol to protect a db/db mouse model of type 2 diabetes against altered metabolic abnormalities and proinflammatory markers via activation of PPARgamma transcription. Eight-week-old db/db mice were treated with cilostazol or rosiglitazone for 12 days. Cilostazol significantly decreased plasma glucose and triglyceride levels, as did rosiglitazone, a PPARgamma agonist. Elevated plasma insulin and resistin levels were significantly decreased by cilostazol, and decreased adiponectin mRNA expression was elevated along with increased plasma adiponectin. Cilostazol significantly increased both adipocyte fatty acid binding protein and fatty acid transport protein-1 mRNA expressions with increased glucose transport 4 in the adipose tissue. Cilostazol and rosiglitazone significantly suppressed proinflammatory markers (superoxide, tumor necrosis factor-alpha, and vascular cell adhesion molecule-1) in the carotid artery of db/db mice. In an in vitro study with 3T3-L1 fibroblasts, cilostazol significantly increased PPARgamma transcription activity, as did rosiglitazone. The transcription activity stimulated by cilostazol was attenuated by KT5720 [(9R,10S,12S)-2,3,9,10,11,12-hexahydro-10-hydroxy-9-methyl-1-oxo-9, 12-epoxy-1H-diindolo[1,2,3-fg:3',2',1'-kl]pyrrolo [3,4-I][1,6]-benzodiazocine-10-carboxylic acid hexyl ester], a cAMP-dependent protein kinase inhibitor, and GW9662 (2-chloro-5-nitrobenzanilide), an antagonist of PPARgamma activity, indicative of implication of the phosphatidylinositol 3-kinase/Akt signal pathway. These results suggest that cilostazol may improve insulin sensitivity along with anti-inflammatory effects in type 2 diabetic patients via activation of both cAMP-dependent protein kinase and PPARgamma transcription.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , PPAR gamma/biosynthesis , Phosphodiesterase Inhibitors/pharmacology , Tetrazoles/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Vascular Cell Adhesion Molecule-1/biosynthesis , 3T3-L1 Cells , Adiponectin/blood , Adiponectin/genetics , Adipose Tissue/drug effects , Adipose Tissue/immunology , Adipose Tissue/metabolism , Animals , Biomarkers/analysis , Blood Glucose/metabolism , Body Weight/drug effects , Cilostazol , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/immunology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression/drug effects , Glucose Transporter Type 4/biosynthesis , Insulin/blood , Male , Mice , Mice, Inbred C57BL , Phosphodiesterase Inhibitors/administration & dosage , Phosphodiesterase Inhibitors/therapeutic use , Receptors, Leptin/deficiency , Tetrazoles/administration & dosage , Tetrazoles/therapeutic use , Transcriptional Activation/drug effects , Triglycerides/blood , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology , Vascular Cell Adhesion Molecule-1/immunology
15.
J Vasc Res ; 46(1): 36-44, 2009.
Article in English | MEDLINE | ID: mdl-18515973

ABSTRACT

This study investigated the effects of 7-ketocholesterol on interleukin (IL)-6 expression in vascular smooth muscle cells (VSMC). Among the 7 IL examined, only IL-6 transcript was increased by 7-ketocholesterol treatment in human aorta smooth muscle cells. IL-6 transcripts increased up to 24 h after treatment with 7-ketocholesterol, and this effect was profoundly repressed by treatment with p38 MAPK inhibitors and to a lesser extent JNK inhibitors. 7alpha-Hydroxycholesterol, 27-hydroxycholesterol or cholesterol, however, did not induce IL-6 expression. Mechanisms of IL-6 induction by 7-ketocholesterol were investigated in comparison with tumor necrosis factor (TNF)-alpha. Whereas TNF-alpha activated IL-6 promoter, which was impaired by p38 MAPK inhibitors or by mutation in the NF-kappaB-binding site within the promoter region, 7-ketocholesterol did not affect IL-6 promoter activity. Instead, this oxysterol slowed degradation of IL-6 mRNA and increased the amount of cytoplasmic HuR. 7-ketocholesterol significantly increased the amount of intracellular IL-6 protein in the presence of brefeldin A. 7-Ketocholesterol also enhanced IL-6 release from VSMC. IL-6 release by 7-ketocholesterol, although significant, was not as remarkable as that induced by TNF-alpha. These data suggest that 7-ketocholesterol upregulates IL-6 via mechanisms distinct from TNF-alpha and contributes to the intra- and extracellular IL-6 deposits within the vasculature.


Subject(s)
Interleukin-6/metabolism , Ketocholesterols/pharmacology , Muscle, Smooth, Vascular/metabolism , Tumor Necrosis Factor-alpha/physiology , Animals , Anthracenes/pharmacology , Brefeldin A/pharmacology , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/physiology , Cells, Cultured , Dactinomycin/pharmacology , ELAV Proteins/metabolism , Humans , Imidazoles/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , NF-kappa B/genetics , NF-kappa B/physiology , Pyridines/pharmacology , RNA, Messenger/metabolism , Rats , Up-Regulation
16.
Vascul Pharmacol ; 50(1-2): 34-9, 2009.
Article in English | MEDLINE | ID: mdl-18824136

ABSTRACT

Despite extensive studies on cellular responses to activation of Toll-like receptor-4 (TLR-4), it is not evident weather its activation affects gene expression of interleukin-8 (IL-8) in vascular smooth muscle cells (VSMCs). Therefore, this study has investigated whether and how TLR-4 influences IL-8 expression in VSMCs. Exposure of aortic smooth muscle cells to TLR-4 agonistic lipopolysaccharide (LPS) not only enhanced release of IL-8 protein but also induced IL-8 gene transcript via promoter activation. The LPS-induced activation of IL-8 promoter was attenuated by dominant-negative MKK1, but not by dominant-negative MKK3. The promoter activation was also impaired by dominant negative CCAAT/enhancer binding protein (C/EBP), IkappaB, and dominant negative c-Jun. In comparison with the mutation of the AP-1 binding site, the mutation of NF-kappaB site and C/EBP binding site in the IL-8 promoter region more significantly impaired the promoter activation. Moreover, both promoter activity and release of IL-8 were inhibited by U0126 and curcumin, but not by SB202190, epigallocatechin 3-gallate and resveratrol. The present study reports that TLR-4-agonistic LPS upregulates IL-8 at the transcriptional and post-translational level in VSMCs, and that ERK1/2, NF-kappaB, and C/EBP play major roles in the upregulation of IL-8.


Subject(s)
Interleukin-8/metabolism , Muscle, Smooth, Vascular/metabolism , Toll-Like Receptor 4/metabolism , Aorta/cytology , Aorta/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cells, Cultured , Humans , Interleukin-8/genetics , Lipopolysaccharides/pharmacology , NF-kappa B/genetics , Promoter Regions, Genetic , RNA Processing, Post-Transcriptional/drug effects , RNA, Messenger/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/agonists , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , Up-Regulation/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
17.
FEBS Lett ; 581(22): 4189-94, 2007 Sep 04.
Article in English | MEDLINE | ID: mdl-17692316

ABSTRACT

To understand the role of adventitial fibroblasts (AF) in diabetic vascular diseases, the importance of high glucose (HG, 25mM) on matrix metalloproteinase-2 (MMP-2) production in AF was determined. HG enhanced mRNA, protein and gelatinolytic activity of MMP-2. The enhanced MMP-2 activity was significantly attenuated not only by a PI3K inhibitor but also by an Akt inhibitor. These HG-induced MMP-2 responses were markedly reduced in Akt1-deficient (1KO) cells. The diminished HG-induced MMP-2 responses were completely restored by re-expression of Akt1. Both the reporter activity and electrophoretic mobility shift assay for activator protein-1 and nuclear factor-kappa B (NF-kappaB) were enhanced by HG, but NF-kappaB were not increased in 1KO cells. Furthermore, HG-induced MMP-2 responses were markedly suppressed by NF-kappaB decoy oligodeoxynucleotides. Based on these results, it is suggested that HG augments MMP-2 production via PI3K/Akt1/NF-kappaB pathway.


Subject(s)
Connective Tissue/enzymology , Fibroblasts/enzymology , Glucose/pharmacology , Matrix Metalloproteinase 2/biosynthesis , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Connective Tissue/drug effects , Enzyme Activation/drug effects , Enzyme Induction/drug effects , Fibroblasts/drug effects , Isoenzymes/metabolism , Matrix Metalloproteinase 2/genetics , Mice , Oligodeoxyribonucleotides/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Signal Transduction/drug effects , Time Factors , Transcription Factors/metabolism , Transcription, Genetic/drug effects
18.
Brain Res ; 1157: 112-20, 2007 Jul 09.
Article in English | MEDLINE | ID: mdl-17521620

ABSTRACT

In the present study, we assessed the beneficial synergistic effects of concurrent treatment with low doses of cilostazol and probucol against focal cerebral ischemic infarct in rats. The ischemic infarct induced by 2-h occlusion of middle cerebral artery (MCA) and 22-h reperfusion was significantly reduced in rat brain that received cilostazol (20 mg/kg) and probucol (30 mg/kg) twice together with prominent improvement of neurological function compared to the effect of cilostazol or probucol monotherapy. Increased myeloperoxidase activity, a marker of neutrophil infiltration, observed in the penumbral zone of vehicle-treated brain was more significantly reduced by cilostazol plus probucol in combination. Increased superoxide-, nitrotyrosine (a marker of peroxynitrite)-, poly(ADP-ribose) [a marker for poly(ADP-ribose) polymerase activity]-, and cleaved caspase-3-positive cells (a proapoptotic marker) in the vehicle sample were significantly attenuated by the combination therapy, while individual treatment with low dose of cilostazol or probucol showed a marginal effect. Taken together, it is suggested that the neuroprotective potentials of combination therapy with low doses of cilostazol plus probucol may provide beneficial therapeutic intervention in reducing the focal cerebral ischemic infarct in rats.


Subject(s)
Brain Ischemia/drug therapy , Cerebral Infarction/drug therapy , Probucol/pharmacology , Telencephalon/drug effects , Tetrazoles/pharmacology , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Apoptosis/drug effects , Apoptosis/physiology , Biomarkers/analysis , Biomarkers/metabolism , Brain Ischemia/metabolism , Brain Ischemia/physiopathology , Caspase 3/analysis , Caspase 3/metabolism , Cerebral Infarction/metabolism , Cerebral Infarction/physiopathology , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/physiology , Cilostazol , Dose-Response Relationship, Drug , Drug Combinations , Drug Synergism , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/physiopathology , Male , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Peroxidase/analysis , Peroxidase/metabolism , Poly(ADP-ribose) Polymerases/analysis , Poly(ADP-ribose) Polymerases/metabolism , Probucol/therapeutic use , Rats , Rats, Sprague-Dawley , Superoxides/antagonists & inhibitors , Superoxides/metabolism , Telencephalon/metabolism , Telencephalon/physiopathology , Tetrazoles/therapeutic use , Treatment Outcome , Tyrosine/analogs & derivatives , Tyrosine/analysis , Tyrosine/metabolism
19.
Biomol Ther (Seoul) ; 23(1): 84-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25593648

ABSTRACT

We investigated the question of whether 7-oxygenated cholesterol derivatives could affect inflammatory and/or immune responses in atherosclerosis by examining their effects on expression of IL-23 in monocytic cells. 7α-Hydroxycholesterol (7αOHChol) induced transcription of the TLR6 gene and elevated the level of cell surface TLR6 protein in THP-1 monocytic cells. Addition of an agonist of TLR6, FSL-1, to TLR6-expressing cells by treatment with 7αOHChol resulted in enhanced production of IL-23 and transcription of genes encoding the IL-23 subunit α (p19) and the IL-12 subunit ß (p40). However, treatment with 7-ketocholesterol (7K) and 7ß-hydroxycholesterol (7ßOHChol) did not affect TLR6 expression, and addition of FSL-1 to cells treated with either 7K or 7ßOHChol did not influence transcription of the genes. Pharmacological inhibition of ERK, Akt, or PI3K resulted in attenuated transcription of TLR6 induced by 7αOHChol as well as secretion of IL-23 enhanced by 7αOHChol plus FSL-1. Inhibition of p38 MAPK or JNK resulted in attenuated secretion of IL-23. These results indicate that a certain type of 7-oxygenated cholesterol like 7αOHChol can elicit TLR6-mediated expression of IL-23 by monocytic cells via PI3K/Akt and MAPKs pathways.

SELECTION OF CITATIONS
SEARCH DETAIL