1
|
Njikan S, Manning AJ, Ovechkina Y, Awasthi D, Parish T. High content, high-throughput screening for small molecule inducers of NF-κB translocation. PLoS One 2018; 13:e0199966. [PMID: 29953522 PMCID: PMC6023200 DOI: 10.1371/journal.pone.0199966] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 06/18/2018] [Indexed: 12/22/2022] Open
Abstract
NF-κB is an important mediator of immune activity and its activation is essential in mounting immune response to pathogens. Here, we describe the optimization and implementation of a high-throughput screening platform that utilizes high content imaging and analysis to monitor NF-κB nuclear translocation. We screened 38,991 compounds from three different small molecule libraries and identified 103 compound as hits; 31% of these were active in a dose response assay. Several of the molecules lacked cytotoxicity or had a selectivity index of more than 2-fold. Our image-based approach provides an important first step towards identifying small molecules with immunomodulatory activity.
Collapse
Affiliation(s)
- Samuel Njikan
- TB Discovery Research, Infectious Disease Research Institute, Seattle, WA, United States of America
| | - Alyssa J. Manning
- TB Discovery Research, Infectious Disease Research Institute, Seattle, WA, United States of America
| | - Yulia Ovechkina
- TB Discovery Research, Infectious Disease Research Institute, Seattle, WA, United States of America
| | - Divya Awasthi
- TB Discovery Research, Infectious Disease Research Institute, Seattle, WA, United States of America
| | - Tanya Parish
- TB Discovery Research, Infectious Disease Research Institute, Seattle, WA, United States of America
- * E-mail:
| |
Collapse
|
2
|
Tiangco DA, Halcomb S, Lattanzio FA, Hargrave BY. 3,4-Methylenedioxymethamphetamine alters left ventricular function and activates nuclear factor-Kappa B (NF-κB) in a time and dose dependent manner. Int J Mol Sci 2010; 11:4843-63. [PMID: 21614177 PMCID: PMC3100831 DOI: 10.3390/ijms11124743] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 11/04/2010] [Accepted: 11/08/2010] [Indexed: 11/16/2022] Open
Abstract
3,4-Methylenedioxymethamphetamine (MDMA) is an illicit psychoactive drug with cardiovascular effects that have not been fully described. In the current study, we observed the effects of acute MDMA on rabbit left ventricular function. We also observed the effects of MDMA on nuclear factor-kappa B (NF-κB) activity in cultured rat ventricular myocytes (H9c2). In the rabbit, MDMA (2 mg/kg) alone caused a significant increase in heart rate and a significant decrease in the duration of the cardiac cycle. Inhibition of nitric oxide synthase (NOS) by pretreatment with L-NAME (10 mg/kg) alone caused significant dysfunction in heart rate, systolic pressure, diastolic pressure, duration of relaxation, duration of cardiac cycle, and mean left ventricular pressure. Pretreatment with L-NAME followed by treatment with MDMA caused significant dysfunction in additional parameters that were not abnormal upon exposure to either compound in isolation: duration of contraction, inotropy, and pulse pressure. Exposure to 1.0 mM MDMA for 6 h or 2.0 μM MDMA for 12 h caused increased nuclear localization of NF-κB in cultured H9c2 cells. The current results suggest that MDMA is acutely detrimental to heart function and that an intact cardiovascular NOS system is important to help mitigate early sequelae in some functional parameters. The delayed timing of NF-κB activation suggests that this factor may be relevant to MDMA induced cardiomyopathy of later onset.
Collapse
Affiliation(s)
- David A. Tiangco
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA
| | - Sapna Halcomb
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA
| | - Frank A. Lattanzio
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, 23510, USA
| | - Barbara Y. Hargrave
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA
| |
Collapse
|
3
|
Lamers D, Schlich R, Greulich S, Sasson S, Sell H, Eckel J. Oleic acid and adipokines synergize in inducing proliferation and inflammatory signalling in human vascular smooth muscle cells. J Cell Mol Med 2010; 15:1177-88. [PMID: 20518853 PMCID: PMC3822630 DOI: 10.1111/j.1582-4934.2010.01099.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
In the context of obesity, perivascular fat produces various adipokines and releases free fatty acids, which may induce inflammation and proliferation in the vascular wall. In this study we investigated how adipokines, oleic acid (OA) and the combined treatment regulate human vascular smooth muscle cell (hVSMC) proliferation and migration and the underlying signalling pathways. Adipocyte-conditioned media (CM) generated from human adipocytes induces a prominent proliferation and migration of hVSMC. Autocrine action of adiponectin totally abolishes CM-induced proliferation. Furthermore, OA but not palmitic acid induces proliferation of hVSMC. CM itself does not contain fatty acids, but CM in combination with OA markedly enhances proliferation of hVSMC in a synergistic way. Both the nuclear factor (NF)-κB and the mammalian target of rapamycin (mTOR) pathway were synergistically activated under these conditions and found to be essential for hVSMC proliferation. Expression of iNOS and production of nitric oxide was only enhanced by combined treatment inducing a marked release of VEGF. Combination of OA and VEGF induces an additive increase of hVSMC proliferation. We could show that the combination of CM and OA led to a synergistic proliferation of hVSMC. Expression of iNOS and production of nitric oxide were only enhanced under these conditions and were paralleled by a marked release of VEGF. These results suggest that the combined elevated release of fatty acids and adipokines by adipose tissue in obesity might be critically related to hVSMC dysfunction, vascular inflammation and the development of atherosclerosis.
Collapse
Affiliation(s)
- Daniela Lamers
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Düsseldorf, Germany
| | | | | | | | | | | |
Collapse
|
4
|
Patel S, Santani D. Role of NF-κB in the pathogenesis of diabetes and its associated complications. Pharmacol Rep 2009; 61:595-603. [DOI: 10.1016/s1734-1140(09)70111-2] [Citation(s) in RCA: 177] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2008] [Revised: 06/13/2009] [Indexed: 12/15/2022]
|
5
|
Cui R, Tieu B, Recinos A, Tilton RG, Brasier AR. RhoA Mediates Angiotensin II–Induced Phospho-Ser536 Nuclear Factor κB/RelA Subunit Exchange on the Interleukin-6 Promoter in VSMCs. Circ Res 2006; 99:723-30. [PMID: 16960103 DOI: 10.1161/01.res.0000244015.10655.3f] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The vasoconstrictor angiotensin II (Ang II) accelerates atherosclerosis by inducing vascular gene expression programs, producing monocyte recruitment, and vascular remodeling. In vascular smooth muscle cells (VSMCs), Ang II signaling activates interleukin (IL)-6 expression, a cytokine producing acute-phase inflammation, mediated by the transcription factor nuclear factor κB (NF-κB). The classical NF-κB activation pathway involves cytoplasmic-to-nuclear translocation of the potent RelA transactivating subunit; however, because nuclear RelA is present in VSMCs, the mechanism by which NF-κB activity is controlled is incompletely understood. In this study, we focus on early activation steps controlling RelA activation. Although Ang II only weakly induces ≈1.5-fold RelA nuclear translocation, RelA is nevertheless required because short interfering RNA–mediated RelA knockdown inhibits inducible IL-6 expression. We find instead that Ang II stimulation rapidly induces RelA phosphorylation at serine residue 536, a critical regulatory site in its transactivating domain. Chromatin immunoprecipitation assays indicate no significant changes in total RelA binding to the native IL-6 promoter, but an apparent increase in fractional binding of phospho-Ser536 RelA. Inactivation of RhoA by treatment with
Clostridium botulinum
exoenzyme C3 exotoxin or expression of dominant negative RhoA blocks Ang II–inducible RelA Ser536 phosphorylation and IL-6 expression. Finally, enhanced phospho-Ser536 RelA formation in the aortae of rats chronically infused with Ang II was observed. Together, these data indicate a novel mechanism for Ang II–induced NF-κB activation in VSMCs, mediated by RhoA-induced phospho-Ser536 RelA formation, IL-6 expression, and vascular inflammation.
Collapse
Affiliation(s)
- Ruwen Cui
- Department of Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA
| | | | | | | | | |
Collapse
|
6
|
Isoda K, Young JL, Zirlik A, MacFarlane LA, Tsuboi N, Gerdes N, Schönbeck U, Libby P. Metformin inhibits proinflammatory responses and nuclear factor-kappaB in human vascular wall cells. Arterioscler Thromb Vasc Biol 2005; 26:611-7. [PMID: 16385087 DOI: 10.1161/01.atv.0000201938.78044.75] [Citation(s) in RCA: 378] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Metformin may benefit the macrovascular complications of diabetes independently of its conventional hypoglycemic effects. Accumulating evidence suggests that inflammatory processes participate in type 2 diabetes and its atherothrombotic manifestations. Therefore, this study examined the potential action of metformin as an inhibitor of pro-inflammatory responses in human vascular smooth muscle cells (SMCs), macrophages (Mphis), and endothelial cells (ECs). METHODS AND RESULTS Metformin dose-dependently inhibited IL-1beta-induced release of the pro-inflammatory cytokines IL-6 and IL-8 in ECs, SMCs, and Mphis. Investigation of potential signaling pathways demonstrated that metformin diminished IL-1beta-induced activation and nuclear translocation of nuclear factor-kappa B (NF-kappaB) in SMCs. Furthermore, metformin suppressed IL-1beta-induced activation of the pro-inflammatory phosphokinases Akt, p38, and Erk, but did not affect PI3 kinase (PI3K) activity. To address the significance of the anti-inflammatory effects of a therapeutically relevant plasma concentration of metformin (20 micromol/L), we conducted experiments in ECs treated with high glucose. Pretreatment with metformin also decreased phosphorylation of Akt and protein kinase C (PKC) in ECs under these conditions. CONCLUSIONS These data suggest that metformin can exert a direct vascular anti-inflammatory effect by inhibiting NF-kappaB through blockade of the PI3K-Akt pathway. The novel anti-inflammatory actions of metformin may explain in part the apparent clinical reduction by metformin of cardiovascular events not fully attributable to its hypoglycemic action.
Collapse
MESH Headings
- Anti-Inflammatory Agents/pharmacology
- Atherosclerosis/drug therapy
- Atherosclerosis/immunology
- Cell Survival/drug effects
- Cells, Cultured
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/immunology
- Diabetic Angiopathies/drug therapy
- Diabetic Angiopathies/immunology
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/immunology
- Glucose/pharmacology
- Humans
- Hypoglycemic Agents/pharmacology
- Interleukin-1/antagonists & inhibitors
- Interleukin-1/metabolism
- Interleukin-6/metabolism
- Interleukin-8/metabolism
- Macrophages/cytology
- Macrophages/drug effects
- Macrophages/immunology
- Metformin/pharmacology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/immunology
- NF-kappa B/metabolism
- Phosphatidylinositol 3-Kinases/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- Saphenous Vein/cytology
Collapse
Affiliation(s)
- Kikuo Isoda
- Donald W. Reynolds Cardiovascular Clinical Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Omura T, Yoshiyama M, Izumi Y, Kim S, Matsumoto R, Enomoto S, Kusuyama T, Nishiya D, Nakamura Y, Akioka K, Iwao H, Takeuchi K, Yoshikawa J. Involvement of c-Jun NH2 Terminal Kinase and p38MAPK in Rapamycin-Mediated Inhibition of Neointimal Formation in Rat Carotid Arteries. J Cardiovasc Pharmacol 2005; 46:519-25. [PMID: 16160607 DOI: 10.1097/01.fjc.0000179001.00779.a5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Rapamycin-coated stents in coronary artery lesions have recently been shown to be effective in inhibiting neointimal formation. However, little is known about the effects of rapamycin on mitogen-activated protein kinase (MAPK), which is an important signal for neointimal formation. Therefore, we examined the effects of rapamycin on MAPK and transcriptional factors in cultured human coronary artery smooth muscle cells (CASMC) and in balloon-injured rat carotid arteries. METHODS AND RESULTS Activation of ERK, JNK, p38MAPK, AP-1, and NF-kB in coronary artery smooth muscle cells was increased by 2% fetal bovine serum. Ten nmol/L of rapamycin prevented the activation of JNK, p38MAPK, AP-1, and NF-kB (65%, 65%, 67%, and 26% respectively, P<0.01). In an in vivo study, remarkable neointimal formation was observed 14 days after injury. Coating Pluronic gel with 20 and 50 mug rapamycin around the injured artery significantly decreased the intimal area/medial area ratio, compared with vehicle (0.75 vs. 1.2, P<0.01). Rapamycin prevented the increase in activation of JNK, p38MAPK, AP-1, and NF-kB in injured artery (42%, 70%, 75%, and 60% respectively, P<0.05). CONCLUSIONS Neointimal formation after balloon injury is inhibited by rapamycin, which is partially mediated by inhibition of JNK and p38MAPK, followed by AP-1 and NF-kB.
Collapse
MESH Headings
- Animals
- Carotid Artery Injuries/enzymology
- Carotid Artery Injuries/metabolism
- Carotid Artery, Common/drug effects
- Carotid Artery, Common/enzymology
- Carotid Artery, Common/metabolism
- Cell Proliferation/drug effects
- Coronary Vessels/drug effects
- Coronary Vessels/enzymology
- Immunosuppressive Agents/pharmacology
- JNK Mitogen-Activated Protein Kinases/metabolism
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- NF-kappa B/metabolism
- Rats
- Rats, Wistar
- Sirolimus/pharmacology
- Transcription Factor AP-1/metabolism
- Tunica Intima/drug effects
- Tunica Intima/enzymology
- Tunica Intima/metabolism
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Takashi Omura
- Department of Internal Medicine and Cardiology, Osaka City University Medical School, Osaka, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Merino VF, Silva JA, Araújo RC, Avellar MCW, Bascands JL, Schanstra JP, Paiva ACM, Bader M, Pesquero JB. Molecular structure and transcriptional regulation by nuclear factor-κB of the mouse kinin B1 receptor gene. Biol Chem 2005; 386:515-22. [PMID: 16006238 DOI: 10.1515/bc.2005.061] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Kinins are important mediators in cardiovascular homeostasis, inflammation, and nociception. Two kinin receptors have been described, B 1 and B 2 . The B 1 receptor is normally absent in healthy tissues, but is highly induced under pathological conditions. To understand the molecular mechanism of B 1 receptor up-regulation, we determined the mouse B 1 receptor gene structure, isolated and characterized the promoter region and studied its transcriptional regulation. The mouse B 1 receptor gene contains two exons (with the entire coding region located in the second exon) and a TATA-less promoter with multiple transcription start sites. A 7.7-kbp portion of the 5'-flanking region was examined for promoter activity in vascular smooth muscle cells (VSMCs). A minimal 92-bp fragment, located immediately upstream of the transcription start region, exerted basal and lipopolysaccharide (LPS)-inducible transcription activity in the sense and antisense orientation, and was thereby identified as an enhancer element. Nuclear extracts from VSMCs showed basal and LPS-inducible binding activity of nuclear factor (NF)-kappaB at this sequence. B 1 receptor transcription activation in response to LPS was abolished by cotransfection with IkappaBalphaDeltaN, an NF-kappaB repressor. In summary, our results reveal the structure of the mouse B 1 receptor gene and the involvement of NF-kappaB in the inducible mouse kinin B 1 receptor expression under pathological conditions.
Collapse
Affiliation(s)
- Vanessa F Merino
- Department of Biophysics, Universidade Federal de São Paulo, CEP 04023-062 São Paulo, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Wang L, MacDonald RC. Effects of Microtubule-Depolymerizing Agents on the Transfection of Cultured Vascular Smooth Muscle Cells: Enhanced Expression with Free Drug and Especially with Drug–Gene Lipoplexes. Mol Ther 2004; 9:729-37. [PMID: 15120334 DOI: 10.1016/j.ymthe.2004.02.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2003] [Accepted: 02/10/2004] [Indexed: 10/26/2022] Open
Abstract
The microtubule-depolymerizing agents colchicine, vinblastine (VB), vincristine, nocodazole, and podophyllotoxin were found to increase dramatically the transfection of cationic phospholipid-DNA (CMV-beta-gal) complexes on cultured vascular smooth muscle cells (VSMCs). Pretreatment of cells with free colchicine before addition of lipoplexes increased transgene expression both in the presence and in the absence of serum. Free vinblastine had similar effects; however, vinblastine was more effective (approximately 30-fold maximal stimulation) when incorporated into the lipoplexes. Under optimal conditions, vincristine, nocodazole, and podophyllotoxin produced 25- and 39-, 31- and 14-, and 26- and 14-fold increases in the absence and presence of serum, respectively. Taxol, which stabilizes microtubules, had no effect on transfection, but it blocked the positive effect of colchicine. Cytochalasin B, which inhibits microfilament polymerization, had no effect on transgene expression. By fluorescence microscopy, normal lipoplexes colocalized with lysosomes. In contrast, there was little, if any, colocalization of VB lipoplexes with lysosomes. Because depolymerization of microtubules induces NF-kappaB-dependent gene expression, the effects of pyrrolidinedithiocarbamate and Nalpha-p-tosyl-L-lysine chloromethyl ketone, inhibitors of NF-kappaB activation, were tested; inhibition of vinblastine stimulation of transfection was 85 and 66%, respectively. Also, immunofluorescence microscopy showed that vinblastine induced the translocation of NF-kappaB from the cytoplasm to the nucleus. It is concluded that microtubule-depolymerizing agents, especially when incorporated into lipoplexes, dramatically increase transfection of VSMCs, probably by two mechanisms: (i) inhibition of transport of lipoplexes to lysosomes and (ii) activation of transcription (via NF-kappaB). There have been some reports on the use of pharmaceutical agents to enhance gene expression, but generally these have involved separate applications of drug and gene. The ability to deliver a drug and a gene in a single therapeutic formulation could have significant clinical implications.
Collapse
Affiliation(s)
- Li Wang
- Department of Biochemistry, Molecular Biology and Cell Biology, Northwestern University, Evanston, IL 60208, USA
| | | |
Collapse
|
10
|
Ferri N, Garton KJ, Raines EW. An NF-kappaB-dependent transcriptional program is required for collagen remodeling by human smooth muscle cells. J Biol Chem 2003; 278:19757-64. [PMID: 12649281 DOI: 10.1074/jbc.m212714200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Although remodeling of vessels can dramatically alter lumen diameter and clinical sequelae, the molecular mechanisms regulating extracellular matrix turnover and remodeling are still not well understood. To investigate these processes in human smooth muscle, we have compared their culture on monomer and polymerized collagen gels, conditions that mimic some of the features of injured and normal vessels, respectively. We show that culture on polymerized, but not monomer, collagen leads to the activation of the transcription factor NF-kappaB through phosphorylation and degradation of its inhibitor, IkappaBalpha. Coincident with NF-kappaB activation, expression of MMP1, MMP2, and alpha2 integrin increases on polymerized collagen. Specific inhibition of NF-kappaB by retroviral overexpression of wild-type IkappaBalpha or phosphorylation-resistant, IkappaBalpha-stabilized mutant (IkappaBalphaSer32,36/Ala) reverses the increases in MMP1 and alpha2beta1 on polymerized collagen and decreases collagen gel contraction and degradation. However, forced overexpression of alpha2beta1 integrin or MMP1 in smooth muscle cells expressing IkappaBalphaSer32,36/Ala rescues their ability to contract collagen gels. Thus, polymerized collagen induces NF-kappaB-dependent expression of MMP1 and alpha2beta1 integrin, that are required for smooth muscle extracellular matrix remodeling.
Collapse
Affiliation(s)
- Nicola Ferri
- Department of Pathology, University of Washington School of Medicine, Seattle 98104, USA
| | | | | |
Collapse
|
11
|
Hofmann CS, Sonenshein GE. Green tea polyphenol epigallocatechin-3 gallate induces apoptosis of proliferating vascular smooth muscle cells via activation of p53. FASEB J 2003; 17:702-4. [PMID: 12586742 DOI: 10.1096/fj.02-0665fje] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Green tea polyphenols (GTPs), which possess antioxidant properties, have been shown to inhibit the development of atherosclerotic lesions. Epigallocatechin-3-gallate (EGCG), the most abundant GTP, displays antiproliferative effects in a variety of cell types. Here, we examined the effects of GTPs on aortic smooth muscle cell (SMC) proliferation. Treatment with a GTP mixture or EGCG at a dose of 40 to 50 microg/ml slowed SMC growth, while at a higher dose of 80 microg/ml EGCG also induced cell death as judged by TUNEL assay. Apoptosis was mainly observed in proliferating SMCs in subconfluent cultures; whereas at higher confluency, cell viability was largely unaffected. Treatment with 80 microg/ml EGCG induced the tumor suppressor p53, which was functional as judged by activation of the target cyclin-dependent kinase inhibitor p21CIP1. Inhibition of p53 activity with a dominant negative mutant reduced cell death. The increase in p53 protein was due to increased stability. EGCG also induced functional nuclear factor-kappaB (NF-kappaB) complexes, and inhibition of this activity reduced the extent of cell death. Thus, EGCG inhibits growth and induces death of SMCs in a p53- and NF-kappaB-dependent manner. These results provide evidence for a new molecular mechanism whereby green tea polyphenols inhibit SMC proliferation and function to prevent the development of atherosclerosis.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Apoptosis/genetics
- Blotting, Northern
- Catechin/analogs & derivatives
- Catechin/pharmacology
- Cattle
- Cell Division/drug effects
- Cyclin-Dependent Kinase Inhibitor p21
- Cyclins/genetics
- Cyclins/metabolism
- Gene Expression Regulation/drug effects
- Immunoblotting
- In Situ Nick-End Labeling
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- NF-kappa B/metabolism
- RNA, Messenger/drug effects
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Tea/chemistry
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
Collapse
Affiliation(s)
- Claudia S Hofmann
- Department of Biochemistry, Boston University School of Medicine, Massachusetts 02118, USA
| | | |
Collapse
|
12
|
Yamasaki K, Asai T, Shimizu M, Aoki M, Hashiya N, Sakonjo H, Makino H, Kaneda Y, Ogihara T, Morishita R. Inhibition of NFkappaB activation using cis-element 'decoy' of NFkappaB binding site reduces neointimal formation in porcine balloon-injured coronary artery model. Gene Ther 2003; 10:356-64. [PMID: 12595894 DOI: 10.1038/sj.gt.3301875] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Application of DNA technology to regulate the transcription of disease-related genes has important therapeutic potential. The transcription factor NFkappaB plays a pivotal role in the transactivation of inflammatory and adhesion molecule genes, leading to vascular lesion formation. Double-stranded DNA with high affinity for NFkappaB may be introduced as 'decoy' cis elements to bind NFkappaB and block the activation of genes mediating inflammation, resulting in effective drugs for treating intimal hyperplasia. In this study, we tested the feasibility of NFkappaB decoy therapy to treat neointimal formation in a porcine coronary artery balloon injury model as a pre-clinical study. An angioplasty catheter was introduced into the left anterior descending coronary artery of the pig to cause vascular injury. First, we tested the feasibility of transfection of FITC-labeled NFkappaB decoy ODN using a hydrogel balloon catheter. Fluorescence due to NFkappaB decoy ODN could be detected throughout the medial layer. Therefore, we transfected NFkappaB decoy ODN into the balloon-injured LAD using a hydrogel catheter. Histological evaluation demonstrated that the neointimal area in the balloon-injured artery was significantly reduced by NFkappaB decoy ODN as compared to scrambled decoy ODN at 1 week after single transfection, accompanied by a significant reduction in PCNA-positive stained cells (P < 0.01). Interestingly, the reduction of ICAM-positive staining was observed, accompanied by the inhibition of migration of macrophages. Of importance, intravascular ultrasound (IVUS) confirmed that neointimal area in the balloon-injured artery was significantly reduced by NFkappaB decoy ODN at 4 weeks after transfection (P < 0.01). Interestingly, the inhibition of neointimal area was only limited to the lesion transfected with NFkappaB decoy ODN, while other lesions without NFkappaB decoy ODN demonstrated a marked increase in neointimal formation. Here, we report the successful in vivo transfer of NFkappaB decoy ODN using a hydrogel catheter to inhibit vascular lesion formation in balloon-injured porcine coronary artery.
Collapse
Affiliation(s)
- K Yamasaki
- Department of Geriatric Medicine, Osaka University Medical School, Suita, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Zhang Y, Sun S, Wang Z, Thompson A, Kaluzhny Y, Zimmet J, Ravid K. Signaling by the Mpl receptor involves IKK and NF-kappaB. J Cell Biochem 2002; 85:523-35. [PMID: 11967992 DOI: 10.1002/jcb.10141] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Binding of tumor necrosis factor-alpha (TNF-alpha) to its receptor activates IKK complex, which leads to inducement of NF-kappaB activity. Here we report that activation of Mpl ligand is also linked to IKK and NF-kappaB activity. Mpl ligand, also known as thrombopoietin (TPO) or megakaryocyte growth and development factor (MGDF), induces megakaryocyte differentiation and inhibition of mitotic proliferation, followed by induction of polyploidization and fragmentation into platelets. The latter process is often observed in megakaryocytes undergoing apoptosis. Treatment of a Mpl ligand-responding megakaryocytic cell line with this cytokine led to an immediate, transient increase in IKK activity followed by a profound decrease in this kinase activity over time. This decrease was not due to an effect on the levels of the IKK regulatory components IKKalpha and IKKbeta. Proliferating megakaryocytes displayed a constitutive DNA-binding activity of NF-kappaB p50 homodimers and of NF-kappaB p50-p65 heterodimers. As expected, reduced IKK activity in Mpl ligand-treated cells was associated with a significant reduction in NF-kappaB DNA binding activity and in the activity of a NF-kappaB-dependent promoter. Our study is thus the first to identify a constitutive NF-kappaB activity in proliferating megakaryocytes as well as to describe a link between Mpl receptor signaling and IKK and NF-kappaB activities. Since a variety of proliferation-promoting genes and anti-apoptotic mechanisms are activated by NF-kappaB, retaining its low levels would be one potential mechanism by which inhibition of mitotic proliferation is maintained and apoptosis is promoted during late megakaryopoiesis.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Biochemistry, Whitaker Cardiovascular Institute, Cancer Research Center, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
|
15
|
Laurindo FRM, de Souza HP, Pedro MDA, Janiszewski M. Redox aspects of vascular response to injury. Methods Enzymol 2002; 352:432-54. [PMID: 12125370 DOI: 10.1016/s0076-6879(02)52039-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Francisco R M Laurindo
- Vascular Biology and Applied Physiology Laboratories, Heart Institute, University of São Paulo School of Medicine, CEP 05403-000 São Paulo, Brazil
| | | | | | | |
Collapse
|
16
|
Yoshimura S, Morishita R, Hayashi K, Yamamoto K, Nakagami H, Kaneda Y, Sakai N, Ogihara T. Inhibition of intimal hyperplasia after balloon injury in rat carotid artery model using cis-element 'decoy' of nuclear factor-kappaB binding site as a novel molecular strategy. Gene Ther 2001; 8:1635-42. [PMID: 11895002 DOI: 10.1038/sj.gt.3301566] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The transcription factor, NFkB, plays a pivotal role in the coordinated transactivation of cytokine and adhesion molecule genes involved in atherosclerosis and lesion formation after vascular injury. We hypothesized that synthetic double-stranded DNA with high affinity for NFkB may be introduced as a 'decoy' cis element to bind the transcription factor, and block gene activation, resulting in an effective therapeutic agent for treating intimal hyperplasia. In vivo transfection of NFkB decoy oligodeoxynucleotides (ODN) into balloon-injured rat carotid artery resulted in the inhibition of neointimal formation at 14 days after injury as compared with vessels transfected with scrambled ODN (P < 0.01). It is of importance to note that in the vessels transfected with NFkB decoy ODN, the expression of p53, a pro-apoptotic gene, was upregulated in neointimal area, followed by increased apoptosis at 14 days. In addition, gene expression of ICAM-1 and VCAM-1 was markedly decreased in blood vessels transfected with NFkB decoy ODN compared with scrambled ODN, whereas balloon injury induced ICAM and VCAM expression in the neointimal area. More importantly, the migration of macrophages and T-lymphocytes into the neointima and media was significantly inhibited by NFkB decoy ODN as compared with scrambled ODN. Here, we demonstrated that in vivo transfer of NFkB decoy ODN successfully inhibited neointimal formation after balloon injury, accompanied by (1) induction of apoptosis through p53 upregulation, and (2) inhibition of local inflammatory actions through the downregulation of adhesion molecules. These results suggest that decoy treatment against NFkB provides a new therapeutic strategy to inhibit neointimal hyperplasia after angioplasty.
Collapse
Affiliation(s)
- S Yoshimura
- Department of Neurosurgery, Gifu University School of Medicine, Japan
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Watanabe T, Yoshizumi M, Akishita M, Eto M, Toba K, Hashimoto M, Nagano K, Liang YQ, Ohike Y, Iijima K, Sudoh N, Kim S, Nakaoka T, Yamashita N, Ako J, Ouchi Y. Induction of nuclear orphan receptor NGFI-B gene and apoptosis in rat vascular smooth muscle cells treated with pyrrolidinedithiocarbamate. Arterioscler Thromb Vasc Biol 2001; 21:1738-44. [PMID: 11701459 DOI: 10.1161/hq1101.098550] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
NGFI-B is one of the orphan nuclear receptors, and its gene is implicated in the apoptosis of T cells. The aim of this study was to investigate the expression and the role of NGFI-B in vascular smooth muscle cells (VSMCs). Pyrrolidinedithiocarbamate (PDTC) is a modulator of an oxidative state and is reported to induce apoptosis only when the density of VSMCs is low. Under low VSMC density (10 000 cells/cm(2)), addition of PDTC (0.1 to 10 micromol/L) caused apoptosis of VSMCs, which was confirmed by Hoechst 33258 staining under fluorescence microscopy. At low VSMC density, expression of NGFI-B mRNA was induced 1 hour after the addition of PDTC, peaking at 6 hours, and persisted for up to 12 hours. The protein level of NGFI-B was increased 4 hours after PDTC addition and persisted for up to 12 hours. Under low VSMC density, PDTC-induced expression of NGFI-B mRNA was correlated with the magnitude of apoptosis, which was quantified by enzyme immunoassay for histone-associated DNA fragments. In contrast, when the density of VSMCs was high (50 000 cells/cm(2)), PDTC did not induce apoptosis, and the expression of NGFI-B was only transient. This transient expression pattern was also seen when VSMCs were treated with phorbol ester, calcium ionophore, hydrogen peroxide, or angiotensin II, even at low cell density. We next investigated whether the NGFI-B gene may act as a transcription factor under treatment with PDTC by measuring the promoter activity of luciferase reporter plasmids that contained typical NGFI-B-responsive elements. The PDTC-induced transcriptional activity of NGFI-B was 2-fold higher at low cell density than at high cell density. These data demonstrate that NGFI-B can be induced in VSMCs and suggest that NGFI-B may play a role in PDTC-induced VSMC apoptosis.
Collapse
MESH Headings
- Animals
- Antioxidants/pharmacology
- Apoptosis
- Cell Culture Techniques/methods
- Cell Nucleus/ultrastructure
- Cells, Cultured
- DNA-Binding Proteins/biosynthesis
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/physiology
- Kinetics
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/ultrastructure
- Nuclear Receptor Subfamily 4, Group A, Member 1
- Pyrrolidines/pharmacology
- RNA, Messenger/biosynthesis
- Rats
- Rats, Sprague-Dawley
- Receptors, Cytoplasmic and Nuclear
- Receptors, Steroid
- Response Elements
- Thiocarbamates/pharmacology
- Transcription Factors/biosynthesis
- Transcription Factors/genetics
- Transcription Factors/physiology
- Transcriptional Activation
- Transfection
Collapse
Affiliation(s)
- T Watanabe
- Department of Geriatric Medicine, Graduate School of Medicine, University of Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
Nuclear factor kappa-B (NFkappaB), a redox-sensitive transcription factor regulating a battery of inflammatory genes, has been indicated to play a role in the development of numerous pathological states. Activation of NFkappaB induces gene programs leading to transcription of factors that promote inflammation, such as leukocyte adhesion molecules, cytokines, and chemokines, although some few substances with possible anti-inflammatory effects are also NFkappaB regulated. The present article reviews basic regulation of NFkappaB and its activation, cell biological effects of NFkappaB activation and the role of NFkappaB in apoptosis. Evidence involving NFkappaB as a key factor in the pathophysiology of ischemia-reperfusion injury and heart failure is discussed. Although activation of NFkappaB induces pro-inflammatory genes, it has lately been indicated that the transcription factor is involved in the signaling of endogenous myocardial protection evoked by ischemic preconditioning. A possible role of NFkappaB in the development of atherosclerosis and unstable coronary syndromes is discussed. Nuclear factor kappa-B may be a new therapeutic target for myocardial protection.
Collapse
Affiliation(s)
- G Valen
- Crafoord Laboratory of Experimental Surgery, Karolinska Hospital, Stockholm, Sweden.
| | | | | |
Collapse
|
19
|
Abstract
Normal embryonic development, tissue differentiation and repair in the eukaryote requires a tightly regulated apoptosis, or programmed cell death. Apoptosis also plays an essential role in different pathological processes including atherosclerosis, in which it affects all cell types in the atherosclerotic lesion, including endothelial cells, vascular smooth muscle cells, and macrophages. During atherosclerosis progression, pro- and anti-apoptotic signals abound in the evolving lesion. Apoptosis limits the number of a particular cell type that accumulates in the lesion and slows down the overall progression of the lesion. On the other hand, it contributes to the production of unstable plaques. Many pharmacological agents used to treat cardiovascular and lipid disorders have pro- or/and anti-apoptotic effects. Pharmaceuticals that modulate apoptosis in specific types of cell can potentially serve as anti-atherogenic agents. However, to develop agents for clinical use requires a thorough knowledge of the pathophysiology of apoptosis in atheromatous lesions, a highly cell-specific process. Here we review our current understanding of the process to provide a background for future pharmacological research in the area.
Collapse
Affiliation(s)
- N V Guevara
- Department of Physical Science, University of Texas at Brownsville, Brownsville, TX 78520, USA
| | | | | |
Collapse
|
20
|
McLaughlin R, Kelly CJ, Kay E, Bouchier-Hayes D. The role of apoptotic cell death in cardiovascular disease. Ir J Med Sci 2001; 170:132-40. [PMID: 11491050 PMCID: PMC7102203 DOI: 10.1007/bf03168827] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Programmed cell death, or apoptosis, is a distinct, managed form of cell death. It is fundamentally different from necrosis. It is a genetically controlled, energy-dependent method of cellular deletion without inflammation. In the cardiovascular system, apoptosis occurs as a primary and secondary event in disease pathogenesis. This review addresses our current understanding of the initiation, propagation and significance of apoptosis in the cardiovascular system, as well as assessing therapeutic potentials arising therefrom. METHODS A Medline search was performed and relevant publications reviewed. Further articles were obtained from the references of these publications. RESULTS and conclusions Apoptotic cell death is a key element in the pathogenesis and progression of ischaemia-reperfusion (IR) injury, cardiac failure, myocardial infarction, atherosclerosis, endothelial dysfunction and the clinical syndromes which these situations produce. Our increased understanding of the role of apoptosis in the pathogenesis of cardiovascular disease offers potential to develop new therapeutic strategies.
Collapse
Affiliation(s)
- R McLaughlin
- Department of Surgery, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | | | | | | |
Collapse
|
21
|
Persson K, Dean-Mckinney T, Steers WD, Tuttle JB. Activation of the transcription factors nuclear factor-kappaB and activator protein-1 in bladder smooth muscle exposed to outlet obstruction and mechanical stretching. J Urol 2001; 165:633-9. [PMID: 11176450 DOI: 10.1097/00005392-200102000-00086] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE Transcriptional control of bladder genes in response to outlet obstruction, growth factors and mechanical force is poorly understood. We analyzed the effects of bladder obstruction, mechanical stretching and platelet derived growth factor on the activation of the major growth controlling transcription factors nuclear factor-kappaB and activator protein-1. MATERIALS AND METHODS Complete outlet obstruction was created in female rats by proximal urethral ligation and bladders were harvested 3, 6 and 24 hours later, respectively. Bladder cells were grown in culture and stimulated with 10 ng./ml. platelet derived growth factor or 10 cycles per minute of mechanical stretching for 0.5 to 4 hours. Nuclear proteins were high salt extracted and incubated with 32phosphorus double strand oligonucleotides containing a consensus binding sequence for activator protein-1 or nuclear factor-kappaB. The resulting DNA protein complexes were analyzed by electrophoretic mobility shift assay. RESULTS Nuclear extract isolated from obstructed bladders showed intense activator protein-1 binding activity 3, 6 and 24 hours after obstruction as well as increased nuclear factor-kappaB binding activity after 6 and 24 hours. Binding activity was absent or minimal in sham operated rats. Cultured cells exposed to mechanical stretching for 2 and 4 hours showed increased activator protein-1 and nuclear factor-kappaB DNA binding compared with unstretched cells. Likewise stimulation with platelet derived growth factor caused a consistent increase in activator protein-1 and nuclear factor-kappaB binding activity. The binding of nuclear proteins was abolished by a 40-fold excess of an unlabeled specific oligonucleotide but not by excess irrelevant oligonucleotide. Thus, the assays were specific for the factors involved. CONCLUSIONS Bladder obstruction and mechanical stretching cause the formation of activator protein-1 and nuclear factor-kappaB DNA complexes, consistent with a role of these transcription factors in the control of hypertrophy associated gene activation.
Collapse
Affiliation(s)
- K Persson
- Department of Clinical Pharmacology, Lund University Hospital, Lund, Sweden
| | | | | | | |
Collapse
|
22
|
De Caterina R, Bourcier T, Laufs U, La Fata V, Lazzerini G, Neish AS, Libby P, Liao JK. Induction of endothelial-leukocyte interaction by interferon-gamma requires coactivation of nuclear factor-kappaB. Arterioscler Thromb Vasc Biol 2001; 21:227-32. [PMID: 11156857 DOI: 10.1161/01.atv.21.2.227] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
To determine whether nuclear factor (NF)-kappaB is necessary to confer endothelial cell responsiveness to interferon (INF)-gamma in terms of vascular cell adhesion molecule (VCAM)-1 expression and leukocyte adhesion, human endothelial cells were treated with IFN-gamma in the presence of low concentrations (LCs) of interleukin (IL)-1alpha (</=100 pg/mL), which activates NF-kappaB but does not induce VCAM-1 expression. Although IFN-gamma induced major histocompatibility complex class II antigen expression and although a high concentration of IL-1alpha (10 ng/mL) induced leukocyte adhesion and VCAM-1 expression, neither IFN-gamma nor LC IL-1alpha was able to induce VCAM-1 expression or leukocyte adhesion. However, the combination of IFN-gamma and LC IL-1alpha induced VCAM-1 expression and increased leukocyte adhesion (67% and 49% of high-concentration IL-1alpha, respectively). Electrophoretic mobility shift assays and immunoblotting of nuclear extracts showed that IFN-gamma activated signal transducers and activators of transcription (STAT)-1alpha and interferon regulatory factor (IRF)-1 but not NF-kappaB, whereas LC IL-1alpha activated NF-kappaB but not STAT-1alpha or IRF-1. Nuclear run-on studies showed that LC IL-1alpha is necessary but not sufficient for inducing VCAM-1 gene transcription and that the combination of IFN-gamma and LC IL-1alpha is required for full VCAM-1 gene transcription. These findings suggest that factors that activate NF-kappaB can synergize with IFN-gamma in promoting endothelial-leukocyte interaction.
Collapse
Affiliation(s)
- R De Caterina
- Vascular Medicine Unit, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Golovchenko I, Goalstone ML, Watson P, Brownlee M, Draznin B. Hyperinsulinemia enhances transcriptional activity of nuclear factor-kappaB induced by angiotensin II, hyperglycemia, and advanced glycosylation end products in vascular smooth muscle cells. Circ Res 2000; 87:746-52. [PMID: 11055977 DOI: 10.1161/01.res.87.9.746] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Pathogenesis of macrovascular complications of diabetes may involve an activation of the transcription factor nuclear factor-kappaB (NF-kappaB) by hyperglycemia and advanced glycosylation end products (AGEs). Activation of NF-kappaB is believed to be dependent on activation of the Rho family of GTPases. Although the precise mechanism of the Rho-mediated action is not completely understood, posttranslational modification of the Rho proteins by geranylgeranylation is required for their subsequent activation. We observed that in cultured vascular smooth muscle cells (VSMCs), insulin stimulated the activity of geranylgeranyltransferase (GGTase) I and increased the amounts of geranylgeranylated Rho-A from 47% to 60% (P:<0.05). GGTI-286, an inhibitor of GGTase I, blocked both effects of insulin. Increased availability of prenylated Rho-A significantly augmented the abilities of angiotensin II (Ang II), hyperglycemia, and AGEs to activate NF-kappaB, as measured by NF-kappaB response-element luciferase reporter activity. Preincubations of VSMCs with insulin for 24 hours doubled NF-kappaB transactivation by Ang II, hyperglycemia, and AGEs. This priming effect of insulin was completely inhibited by GGTI-286. We demonstrate for the first time, to our knowledge, that insulin potentiates NF-kappaB-dependent transcriptional activity induced by hyperglycemia, AGEs, and Ang II in VSMCs by increasing the activity of GGTase I and the availability of geranylgeranylated Rho-A.
Collapse
Affiliation(s)
- I Golovchenko
- Department of Medicine and Research Service of the Denver VA Medical Center, Denver, Colorado, USA
| | | | | | | | | |
Collapse
|
24
|
Marra DE, Simoncini T, Liao JK. Inhibition of vascular smooth muscle cell proliferation by sodium salicylate mediated by upregulation of p21(Waf1) and p27(Kip1). Circulation 2000; 102:2124-30. [PMID: 11044431 DOI: 10.1161/01.cir.102.17.2124] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Salicylates may have direct vascular effects by mechanisms that are independent of platelet inhibition. METHODS AND RESULTS We investigated the effect of salicylates on vascular smooth muscle cell (SMC) proliferation in response to platelet-derived growth factor (PDGF) in vitro. Salicylate concentrations of 5 and 10 mmol/L inhibited serum- or PDGF-induced SMC cell count and [(3)H]thymidine incorporation by 62% and 81%, respectively. There was no evidence of cellular toxicity or apoptosis as determined by trypan blue exclusion and FACS analyses. Because cell cycle progression is regulated by hyperphosphorylation of the retinoblastoma (Rb) protein, we examined the effects of salicylate on Rb hyperphosphorylation. Treatment with salicylate, but not indomethacin, inhibited nuclear factor-kappaB activation and completely abolished Rb hyperphosphorylation in PDGF-treated SMCs. This effect was associated with a decrease in cyclin-dependent kinase (Cdk)-2 and, to a lesser extent, Cdk-6, but not Cdk-4 activity, without changes in Cdk-2, -4, and -6 and cyclin D and E protein levels. Because Cdk-2 activity is regulated by the Cdk inhibitors p21(Waf1) and p27(Kip1), we studied the effects of salicylate on p21(Waf1) and p27(Kip1) expression. Treatment with salicylate prevented PDGF-induced downregulation of p21(Waf1) and p27(Kip1) but not of the Cdk-4/-6 inhibitor p16(Ink4). CONCLUSIONS These findings indicate that high doses of salicylates inhibit SMC proliferation by cell cycle arrest at the G(1)-S phase and suggest a beneficial role for high-dose salicylates in the treatment of vascular proliferative disorders.
Collapse
Affiliation(s)
- D E Marra
- Cardiovascular Division, Brigham & Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
25
|
Weber C, Erl W. Modulation of vascular cell activation, function, and apoptosis: role of antioxidants and nuclear factor-kappa B. CURRENT TOPICS IN CELLULAR REGULATION 2000; 36:217-35. [PMID: 10842754 DOI: 10.1016/s0070-2137(01)80010-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The activity of NF-kappa B is critically involved in the inflammatory activation of endothelial cells and their adhesiveness and also appears to regulate apoptosis in SMC by coordinating antiapoptotic programs. The activity of NF-kappa B has been revealed within human atheromas or following angioplasty but not in undiseased arteries. Hence, the inhibition of NF-kappa B mobilization by antioxidative or anti-inflammatory agents or by adenoviral I kappa B alpha overexpression, as reviewed herein, may act in concert to suppress endothelial activation and to induce SMC apoptosis. This synergistic concept may be a vasoprotective approach to prevent atherogenesis and restenosis by attenuating inflammatory reactions and SMC proliferation in nascent and progressing atherosclerotic lesions, as well as in developing neointima formations following angioplasty.
Collapse
Affiliation(s)
- C Weber
- Institut für Prophylaxe und Epidemiologie der Kreislaufkrankheiten Ludwig-Maximilians-Universität Munich, Germany
| | | |
Collapse
|
26
|
Sasu S, Beasley D. Essential roles of IkappaB kinases alpha and beta in serum- and IL-1-induced human VSMC proliferation. Am J Physiol Heart Circ Physiol 2000; 278:H1823-31. [PMID: 10843878 DOI: 10.1152/ajpheart.2000.278.6.h1823] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Interleukin-1 (IL-1) is a potent vascular smooth muscle cell (VSMC) mitogen, which can stimulate cells via activation of nuclear factor-kappaB (NF-kappaB) following phosphorylation of its inhibitory subunit (IkappaB). Because the proliferative effect of IL-1 is additive with that of serum, the present studies assessed the role of IkappaB kinases (IKKs) and NF-kappaB in both IL-1- and serum-induced VSMC proliferation. IL-1beta (1 ng/ml) induced marked and persistent NF-kappaB activation in VSMC that was maximal at 1 h and persisted for 3 days. There was a 3-fold increase in DNA synthesis after acute IL-1 exposure (24-96 h) and a 12-fold increase after chronic IL-1 exposure (>7 days). Electrophoretic mobility shift assay and supershift analysis indicated that IL-1-induced NF-kappaB complexes consisted of p65/p50 heterodimers and p50 homodimers. Human saphenous vein smooth muscle cells (HSVSMC) were transiently cotransfected with expression plasmids encoding a dominant negative mutant form of either IKKalpha or IKKbeta, in which K(44) was mutated to A (K44A), and a green fluorescent protein expression plasmid that allows identification of transfected cells. IL-1 induced nuclear localization of p65 in 95% of cells transfected with vector alone but in only 69% and 26% of cells expressing IKKalpha (K44A) or IKKbeta (K44A), respectively. Likewise, proliferation increased 3.2-fold in IL-1-treated HSVSMC which had been transfected with vector alone, but only 2.2- and 1.5-fold proliferation in HSVSMC expressing IKKalpha (K44A) or IKKbeta (K44A), respectively. Although serum activated NF-kappaB transiently, serum-induced proliferation was markedly attenuated in HSVSMC expressing IKKalpha (K44A) and IKKbeta (K44A) compared with HSVSMC transfected with vector alone. The results support an essential role of IKKs in the proliferative response of HSVSMC to IL-1 and to serum.
Collapse
Affiliation(s)
- S Sasu
- Department of Medicine and Tupper Research Institute, New England Medical Center Hospitals, Tufts University School of Medicine, Boston, MA 02111, USA
| | | |
Collapse
|
27
|
Erl W, Weber C, Hansson GK. Pyrrolidine dithiocarbamate-induced apoptosis depends on cell type, density, and the presence of Cu(2+) and Zn(2+). Am J Physiol Cell Physiol 2000; 278:C1116-25. [PMID: 10837339 DOI: 10.1152/ajpcell.2000.278.6.c1116] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Pyrrolidine dithiocarbamate (PDTC) has been found to induce or inhibit apoptosis in different cell types. Here we show that PDTC dose-dependently reduced the viability of rat smooth muscle cells (rSMC), human fibroblasts, and endothelial cells at low but not at high cell density. Endothelial cells were least sensitive, fibroblasts showed a medium sensitivity, and rSMC showed a high sensitivity to PDTC-mediated cell death. An early reduction in the mitochondrial membrane potential indicated a rapid onset of apoptosis in rSMC. Apoptosis was further confirmed by annexin V staining and DNA fragmentation analysis. Gel shift analysis demonstrated increased nuclear factor (NF)-kappaB activity in high-density rSMC compared with low-density cells. NF-kappaB has recently been shown to regulate the induction of anti-apoptotic proteins. Although PDTC is widely used as an inhibitor for NF-kappaB and a radical scavenger, our data show that PDTC rather enhanced NF-kappaB activity and, alone or in combination with menadione, induced oxygen radical generation. Notably, PDTC failed to reduce rSMC viability in medium without Cu(2+) or Zn(2+), and addition of Cu(2+) or Zn(2+) resulted in a dose-dependent increase in PDTC-induced cell death. Addition of both Cu(2+) and Zn(2+) showed synergistic effects. Our results indicate that the induction of apoptosis by PDTC requires Cu(2+) and Zn(2+) and is dependent on cell type and density. Such differential effects may have implications for studies of PDTC as an anti-atherosclerotic or immunomodulatory drug.
Collapse
MESH Headings
- Animals
- Antioxidants/pharmacology
- Aorta
- Apoptosis/drug effects
- Apoptosis/physiology
- Cell Membrane/drug effects
- Cell Membrane/physiology
- Cell Survival/drug effects
- Cells, Cultured
- Copper/pharmacology
- Copper Sulfate/pharmacology
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/physiology
- Fibroblasts/cytology
- Fibroblasts/drug effects
- Fibroblasts/physiology
- Humans
- Intracellular Membranes/drug effects
- Intracellular Membranes/physiology
- Kinetics
- Male
- Mitochondria/drug effects
- Mitochondria/physiology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiology
- Pyrrolidines/pharmacology
- Rats
- Rats, Sprague-Dawley
- Reactive Oxygen Species/physiology
- Thiocarbamates/pharmacology
- Umbilical Veins
- Vitamin K/pharmacology
- Zinc/pharmacology
- Zinc Sulfate/pharmacology
Collapse
Affiliation(s)
- W Erl
- Cardiovascular Research Unit, Karolinska Institute, Stockholm, Sweden.
| | | | | |
Collapse
|
28
|
Souza HP, Souza LC, Anastacio VM, Pereira AC, Junqueira ML, Krieger JE, da Luz PL, Augusto O, Laurindo FR. Vascular oxidant stress early after balloon injury: evidence for increased NAD(P)H oxidoreductase activity. Free Radic Biol Med 2000; 28:1232-42. [PMID: 10889453 DOI: 10.1016/s0891-5849(00)00240-9] [Citation(s) in RCA: 95] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Available evidence for oxidative stress after angioplasty is indirect or ambiguous. We sought to characterize the pattern, time course, and possible sources of free radical generation early after arterial balloon injury. Ex vivo injury performed in arterial rings in buffer with lucigenin yielded a massive oxygen-dependent peak of luminescence that decayed exponentially and was proportional to the degree of injury. Signals for injured vs. control arteries were 207. 1 +/- 17.9 (n = 13) vs 4.1 +/- 0.7 (n = 22) cpm x 10(3)/mg/min (p <. 001). Data obtained with 0.25 mmol/l lucigenin were validated with 0. 005-0.05 mmol/l lucigenin or the novel superoxide-sensitive probe coelenterazine (5 micromol/l). Gentle removal of endothelium prior to injury scarcely affected the amount of luminescence. Lucigenin signals were amplified 5- to 20-fold by exogenous NAD(P)H, and were >85% inhibited by diphenyliodonium (DPI, a flavoenzyme inhibitor). Antagonists of several other potential free radical sources, including xanthine oxidase, nitric oxide synthase, and mitochondrial electron transport, were without effect. Overdistension of intact rabbit iliac arteries in vivo (n = 7) induced 72% fall in intracellular reduced glutathione and 68% increase in oxidized glutathione, so that GSH/GSSG ratio changed from 7.93 +/- 2.14 to 0. 81 +/- 0.16 (p <.005). There was also 28.7% loss of the glutathione pool. Further studies were performed with electron paramagnetic resonance spectroscopy. Rabbit aortas submitted to ex vivo overdistension in the presence of the spin trap DEPMPO (5-diethoxy-phosphoryl-5-methyl-1-pyrroline-N-oxide, 100 mmol/l, n = 5) showed formation of radical adduct spectra, abolished by DPI or superoxide dismutase. Computer simulation indicated a mixture of hydroxyl and carbon-centered radical adducts, likely due to decay of superoxide adduct. Electrical mobility shift assays for NF-kappaB activation were performed in nuclear protein extracts from intact or previously injured rabbit aortas. Balloon injury induced early NF-kappaB activation, which was decreased by DPI. In conclusion, our data show unambiguously that arterial injury induces an immediate profound vascular oxidative stress. Such redox imbalance is likely accounted for by activation of vessel wall NAD(P)H oxidoreductase(s), generating radical species potentially involved in tissue repair.
Collapse
Affiliation(s)
- H P Souza
- Emergency Medicine Research Laboratory, University of São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Kaul S, Cercek B, Rengstrom J, Xu XP, Molloy MD, Dimayuga P, Parikh AK, Fishbein MC, Nilsson J, Rajavashisth TB, Shah PK. Polymeric-based perivascular delivery of a nitric oxide donor inhibits intimal thickening after balloon denudation arterial injury: role of nuclear factor-kappaB. J Am Coll Cardiol 2000; 35:493-501. [PMID: 10676700 DOI: 10.1016/s0735-1097(99)00543-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVES To examine the effect of a polymeric-based periadventitial delivery of a nitric oxide (NO)-releasing diazeniumdiolate, spermine/NO (SPER/NO), on balloon injury-induced neointimal hyperplasia in rat ileofemoral arteries. BACKGROUND Reduced local bioavailability and adverse side effects limit systemic administration of NO to modulate vascular response to injury. METHODS A polylactic-polyglycolic acid polymeric matrix containing 2.5% SPER/NO (w/w) was applied around the injured arteries. Quantitative histomorphometry was performed at day 14, proliferating cell nuclear antigen (PCNA) immunohistochemistry at day 3 to assess effects on smooth muscle proliferation and electrophoretic mobility shift assay to evaluate effects on transcription factor, nuclear factor-kappaB (NF-kappaB). RESULTS Treatment with SPER/NO reduced the intimal area (0.011 +/- 0.009 vs. 0.035 +/- 0.006 mm2 control, p < 0.01) and the intima to media ratio (0.089 +/- 0.062 vs. 0.330 +/- 0.057 control, p < 0.005). Spermine/nitric oxide produced a profound inhibition of PCNA-positive cells (>75%, p < 0.005) and significantly suppressed the injury-induced activation of NF-kappaB. Vascular cyclic guanosine monophosphate (cGMP) levels were elevated after treatment with the SPER/NO (0.28 +/- 0.03 vs. 0.17 +/- 0.02 pmol/mg tissue control, p < 0.01). The inhibitory effects on neointimal proliferation were localized to the site of application of SPER/NO and were not associated with any changes in platelet aggregation or bleeding time. Neither SPER nor polymer alone had any significant effects on any of the variables examined. CONCLUSIONS Polymeric-based perivascular delivery of a NO donor produces a marked localized inhibition of neointimal proliferation in balloon-injured arteries. This phenomenon is associated with suppression of NF-kappaB activation and elevation of the vascular cGMP at the site of injury.
Collapse
Affiliation(s)
- S Kaul
- Burns and Allen Research Institute and the Division of Cardiology, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Hoshi S, Goto M, Koyama N, Nomoto K, Tanaka H. Regulation of vascular smooth muscle cell proliferation by nuclear factor-kappaB and its inhibitor, I-kappaB. J Biol Chem 2000; 275:883-9. [PMID: 10625622 DOI: 10.1074/jbc.275.2.883] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Proliferation of vascular smooth muscle cells (SMC) is a crucial event in the formation of atherosclerotic tissues and is regulated by nuclear transcriptional factors including nuclear factor-kappaB (NF-kappaB). We constructed a reporter gene assay to measure NF-kappaB-dependent transcriptional activity in SMC. Thrombin receptor-activating peptide (TRAP) and basic fibroblast growth factor (bFGF) stimulated SMC proliferation and rapidly enhanced the NF-kappaB transcriptional activity in a dose-dependent manner. 4-Cyano-5,5-bis-(methoxyphenyl)4-pentenoic acid (E5510) significantly inhibited SMC proliferation and also suppressed NF-kappaB transcription stimulated by TRAP and bFGF. In contrast, although tumor necrosis factor (TNF)-alpha activated NF-kappaB transcription, E5510 had no effect on TNF-alpha-induced activation. NF-kappaB was activated after the stimulation of TRAP, bFGF, and TNF-alpha in electrophoretic mobility shift assay, and E5510 suppressed the NF-kappaB activation induced by TRAP and bFGF but not the activation by TNF-alpha. Western blot analysis of I-kappaBalpha and I-kappaBbeta, inhibitors of NF-kappaB, indicated that I-kappaBalpha degradation, rather than I-kappaBbeta degradation, was important in NF-kappaB activation after the stimulation of TRAP and bFGF. PD98059, an inhibitor of extracellular signal-regulated kinase (ERK) kinase, suppressed NF-kappaB transcriptional activity and SMC proliferation. The phosphorylation of ERK1/2 was rapidly induced by TRAP and bFGF but not by TNF-alpha. These results indicate that TRAP and bFGF induced I-kappaB degradation and NF-kappaB activation through a distinct pathway from TNF-alpha and that ERK1/2 may play an important role in NF-kappaB activation induced by TRAP and bFGF.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/cytology
- Aorta, Thoracic/metabolism
- Cell Division/drug effects
- Cells, Cultured
- DNA-Binding Proteins/metabolism
- Fatty Acids, Monounsaturated/pharmacology
- Fibroblast Growth Factor 2/pharmacology
- Flavonoids/pharmacology
- Gene Expression Regulation
- Genes, Reporter
- I-kappa B Proteins
- Kinetics
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3
- Mitogen-Activated Protein Kinases/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- NF-KappaB Inhibitor alpha
- NF-kappa B/antagonists & inhibitors
- NF-kappa B/metabolism
- Peptide Fragments/pharmacology
- Phosphorylation
- Platelet Aggregation Inhibitors/pharmacology
- Rats
- Transcription, Genetic/drug effects
- Tumor Necrosis Factor-alpha/pharmacology
Collapse
Affiliation(s)
- S Hoshi
- Eisai Co. Ltd., Tsukuba Research Laboratories, Tokodai 5-1-3, Tsukuba, Ibaraki 300-2635, Japan.
| | | | | | | | | |
Collapse
|
31
|
Elsharkawy AM, Wright MC, Hay RT, Arthur MJ, Hughes T, Bahr MJ, Degitz K, Mann DA. Persistent activation of nuclear factor-kappaB in cultured rat hepatic stellate cells involves the induction of potentially novel Rel-like factors and prolonged changes in the expression of IkappaB family proteins. Hepatology 1999; 30:761-9. [PMID: 10462383 DOI: 10.1002/hep.510300327] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Rat hepatic stellate cells (HSC) cultured in serum-containing medium underwent a rapid (3-hour) classical induction of p50:p65 and p65:p65 nuclear factor-kappaB (NF-kappaB) dimers. Subsequent culturing was associated with prolonged expression of active p50:p65 and persistent induction of a high-mobility NF-kappaB DNA binding complex consisting of potentially novel Rel-like protein(s). Formation of the latter complex was competed for by specific double-stranded oligonucleotides, was up-regulated by treatment of HSCs with tumor necrosis factor alpha (TNF-alpha), and was maintained at basal levels of expression by a soluble HSC-derived factor. An NF-kappaB-responsive CAT reporter gene was highly active in early cultured HSCs but was also trans-activated at a lower but significant level in longer-term cultured cells and could be completely suppressed by expression of dominant negative IkappaB-alpha. Physiological significance of the lower persistent NF-kappaB activities was also demonstrated by the ability of long-term cultured HSCs to support the activity of the NF-kappaB-dependent human intercellular adhesion molecule-1 (ICAM-1) promoter. Freshly isolated HSCs expressed high levels of IkappaB-alpha and IkappaB-beta. Culture activation was accompanied by a long-term reduction in levels of IkappaB-alpha with no detectable expression in the nuclear fraction of cells, under these conditions p50:p65 was detected in the nucleus. IkappaB-beta expression was transiently reduced and, upon replenishment, was associated with appearance of a lower-mobility IkappaB-beta antibody-reactive species. Bcl3 expression was absent in freshly isolated HSC but was induced during culturing and became a persistent feature of the activated HSC. Inhibition of NF-kappaB DNA binding activity by gliotoxin was associated with increased numbers of apoptotic cells. We suggest that activation of NF-kappaB in cultured HSC is required for expression of specific genes associated with the activated phenotype such as ICAM-1 and may be antiapoptotic for rat HSCs.
Collapse
Affiliation(s)
- A M Elsharkawy
- University of Medicine, Southampton General Hospital, Southampton, England
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Newman WH, Zunzunegui RG, Warejcka DJ, Dalton ML, Castresana MR. A reactive oxygen-generating system activates nuclear factor-kappaB and releases tumor necrosis factor-alpha in coronary smooth muscle cells. J Surg Res 1999; 85:142-7. [PMID: 10383851 DOI: 10.1006/jsre.1999.5668] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND Recently we reported that bacterial lipopolysaccharide (LPS) stimulates release of tumor necrosis factor alpha (TNF-alpha) from porcine coronary arteries and smooth muscle cells cultured from those vessels. It has also been reported that plasma levels of TNF-alpha are elevated after myocardial infarction. Since it is known that the production of reactive oxygen intermediates (ROI) occurs during ischemia and ROI are suggested activators of the nuclear regulatory factor kappaB (NF-kappaB), we tested the hypothesis that release of TNF-alpha from smooth muscle cells could also be stimulated with a ROI-generating system. MATERIALS AND METHODS Smooth muscle cells were isolated from porcine coronary arteries. Confluent cells in 48-well culture dishes were treated for 30 min with 0.003 units/ml xanthine oxidase (XO) and 2 mM hypoxanthine (HX) added to the culture medium. The medium was then removed and the cells were washed three times and fresh medium without HX-XO was added. Then, at 1, 3, and 6 h the medium was removed and analyzed for biologically active TNF-alpha. In other experiments, smooth muscle cells were treated with 20 micrograms/ml LPS for 6 h and aliquots of medium analyzed for TNF-alpha. Untreated cells served as controls. Data were analyzed by two-way ANOVA with repeated measures. Extracts of total cell protein were prepared and activation of NF-kappaB was determined by electrophoretic mobility shift assay. RESULTS Treatment of cells with HX-XO stimulated release of TNF-alpha, which rose to a maximum of 17.5 +/- 1.7 units/mg cell protein at 6 h. This was significantly higher (P < 0. 05) than release stimulated by LPS (10.2 +/- 1.0 units/mg at 6 h) or TNF-alpha detected in the culture medium from untreated control cells (4.2 +/- 0.9 units/mg protein at 6 h). Both HX/XO and LPS activated NF-kappaB. CONCLUSIONS These results support the conclusion that coronary smooth muscle cells are a potential source of TNF-alpha during events that are associated with formation of ROI such as myocardial ischemia.
Collapse
Affiliation(s)
- W H Newman
- Department of Surgery, Mercer University School of Medicine and Medical Center of Central Georgia, Macon, Georgia, 31201, USA
| | | | | | | | | |
Collapse
|
33
|
Huynh TT, Davies MG, Trovato MJ, Barber L, Safi HJ, Hagen PO. Reduction of lipid peroxidation with intraoperative superoxide dismutase treatment decreases intimal hyperplasia in experimental vein grafts. J Surg Res 1999; 84:223-32. [PMID: 10357924 DOI: 10.1006/jsre.1999.5647] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND Vein graft failure is commonly attributed to the development of intimal hyperplastic lesions. Oxidative stress has been implicated in the initiation and progression of atherosclerosis. In this study we examined the effects of local intraoperative treatment with polyethylene glycolated superoxide dismutase (PEG-SOD) on lipid peroxidation and on the development of intimal hyperplasia in experimental vein grafts. MATERIALS AND METHODS Forty-one New Zealand White male rabbits had a right carotid interposition bypass graft using the ipsilateral reversed jugular vein. Sixteen animals received local PEG-SOD (4,100 units) treatment; 9 animals received the polyethylene glycol (PEG) vehicle without SOD; 16 animals were used as controls. Postoperatively, malondialdehyde (MDA, a product of lipid peroxidation) concentration and SOD activity were assessed in 3-day vein grafts by colorimetric spectrophotometry. To determine wall dimensions and vasomotor function, morphometric and isometric tension studies were performed on 28-day vein grafts. RESULTS MDA concentration was increased 5. 7-fold (P < 0.05) in 3-day control vein grafts compared to ungrafted jugular veins. Intraoperative PEG-SOD treatment raised SOD activity 5.0-fold (P < 0.05) and reduced MDA concentration 8-fold (P < 0.05) in 3-day vein grafts compared to controls. At 28 days, intimal thickness was reduced by 35% with PEG-SOD treatment (54 +/- 4 vs 83 +/- 5; P < 0.001) compared to control vein grafts, without a change in medial thickness (77 +/- 4 vs 88 +/- 5; P = ns). The vasomotor functions of 28-day PEG-SOD-treated vein grafts to norepinephrine, serotonin, bradykinin, nitroprusside, and acetylcholine were not significantly changed when compared to controls. Treatment with PEG alone did not significantly alter lipid peroxidation, wall dimensions, or vasomotor function of vein grafts. CONCLUSION This study demonstrates that intraoperative local treatment of vein grafts with PEG-SOD increases SOD activity and decreases lipid peroxidation for at least 3 days, resulting in reduced intimal hyperplasia at 28 days. These findings further implicate oxidative stress in the hyperplastic response of vein grafts and suggest a potential therapeutic role for PEG-SOD in the prevention of vein graft failure.
Collapse
Affiliation(s)
- T T Huynh
- Vascular Biology and Atherosclerosis Research Laboratory, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | | | | | | | | | | |
Collapse
|
34
|
Maret A, Clamens S, Delrieu I, Elhage R, Arnal JF, Bayard F. Expression of the interleukin-6 gene is constitutive and not regulated by estrogen in rat vascular smooth muscle cells in culture. Endocrinology 1999; 140:2876-82. [PMID: 10342880 DOI: 10.1210/endo.140.6.6763] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Vascular smooth muscle cells (SMC) are major constituents of the medial layer of blood vessels and are involved in the development of atherosclerotic plaque. SMC secrete copious IL-6 under basal conditions that can be increased by cytokines such as tumor necrosis factor-alpha and interleukin-1beta (IL-1beta). The goal of our studies was to define the role of estrogen in IL-6 production by SMC. In a first series of experiments, the expression of specific messenger RNAs as well as the production of IL-6 bioactivity by rat SMC in culture could be demonstrated in basal and IL-1-stimulated conditions, but was unaffected by estrogen treatment. Different constructs containing deleted or mutated fragments of the human IL-6 promoter driving luciferase or chloramphenicol acetyltransferase reporter gene were then transiently transfected in these cells. A significant basal activity that was increased 2- to 4-fold after IL-1beta stimulation was observed with the total IL-6 promoter. Deletion analysis indicated that the -158/+11 region containing activator protein-1 and cAMP response element sites was apparently the minimal region of IL-6 promoter to confer both constitutive and IL-1-inducible activities. Site-directed mutagenesis experiments suggest that basal activity is dependent upon the promoter sequence -158 to -112 containing the nuclear factor (NF)-IL6(-153) and Sp1 sites, whereas IL-1beta stimulation would depend on the residual -112 nucleotides containing NF-IL6(-75) and NF-kappaB sites. In contrast to the down-regulation of IL-6 expression by estrogen described in osteoblasts, ethinyl estradiol as well as 17beta-estradiol did not influence stimulated IL-6 activity in our experimental conditions whatever the construct tested, even when either estrogen receptor alpha or beta was overexpressed. Thus, the atheroprotective properties of estrogen are probably not mediated through the regulation of IL-6 production by SMC.
Collapse
Affiliation(s)
- A Maret
- INSERM U-397, Institute Louis Bugnard, Toulouse, France.
| | | | | | | | | | | |
Collapse
|
35
|
Kypreos KE, Marhamati DJ, Sonenshein GE. B-Myb represses trans-activation of the Col5A2 collagen promoter indirectly via inhibition of binding of factors interacting with positive elements within the first exon. Matrix Biol 1999; 18:275-85. [PMID: 10429946 DOI: 10.1016/s0945-053x(99)00023-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
B-myb, a member of the myb gene family, was originally isolated based on its high homology with c-myb in the DNA-binding domain. Previously we showed that B-myb is expressed in bovine vascular smooth muscle cells (SMCs) in a cell cycle-dependent fashion, and inhibits type I collagen gene promoter activity. Here, we have explored its role in regulation of another fibrillar collagen gene, Col5A2, encoding the (alpha2 chain of type V collagen. Ectopic expression of B-Myb decreased alpha 2(V) promoter activity and endogenous alpha 2(V) collagen mRNA levels. The responsive region of the alpha 2(V) collagen gene was localized to a fragment including 100 bp of basal promoter and 150 bp of exon 1 sequences, which contained two CRE-like elements. Binding to these elements increased upon deprivation of serum-growth factors, when expression of the Col5A2 gene is elevated, leading us to test their role despite the failure of excess unlabelled CRE oligonucleotide from the somatostatin gene to successfully compete for binding. Mutation of the elements significantly decreased the basal level of alpha2(V) collagen promoter activity and ablated inhibition by B-Myb. Furthermore, addition of B-Myb-glutathionine S-transferase fusion protein inhibited complex formation. Thus, these results confirm a major role for B-Myb in mediating intracellular signals controlling collagen gene expression in vascular SMCs. A model of indirect repression of the Col5A2 gene by B-Myb, via interaction with a positively-acting matrix regulatory factor, termed MRF-V, is discussed.
Collapse
Affiliation(s)
- K E Kypreos
- Department of Biochemistry, Boston University School of Medicine, MA 02118, USA
| | | | | |
Collapse
|
36
|
Selzman CH, Shames BD, McIntyre RC, Banerjee A, Harken AH. The NFkappaB inhibitory peptide, IkappaBalpha, prevents human vascular smooth muscle proliferation. Ann Thorac Surg 1999; 67:1227-31; discussion 1231-2. [PMID: 10355388 DOI: 10.1016/s0003-4975(99)00252-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Vessel injury results in an inflammatory response characterized by the elaboration of cytokines and growth factors, which ultimately influence vascular smooth muscle cell (VSMC) growth and contribute to atherogenesis. Nuclear factor-kappa B (NFkappaB) is a central transcription factor important in mediating stress and inflammatory-induced signals. We hypothesized that strategies aimed at inhibiting NFkappaB would abrogate mitogen-induced human VSMC proliferation. METHODS Human aortic VSMC were stimulated with basic fibroblast growth factor (FGF) and tumor necrosis factor-alpha (TNF), and proliferation was quantified by a colormetric assay. The influence of NFkappaB on VSMC proliferation was examined by both nonspecific NFkappaB blockade with calpain inhibitor-1 (CI-1) and dexamethasone (Dex) and specific NFkappaB blockade with liposomal delivery of the NFkappaB inhibitory peptide, IkappaBalpha. RESULTS FGF and TNF induced concentration-dependent VSMC proliferation (p < 0.002). Neither CI-1, Dex, nor liposomal IkappaBalpha influenced proliferation of unstimulated VSMC. However, both FGF- and TNF-stimulated VSMC proliferation was inhibited to the level of control with CI-1, Dex, and liposomal IkappaBalpha (p < 0.001). CONCLUSION The mitogenic effect of FGF and TNF on human arterial VSMC may be prevented by inhibiting NFkappaB. Furthermore, liposomal delivery of endogenous inhibitory proteins such as IkappaBalpha may represent a novel, therapeutically accessible method for selective transcriptional suppression in the response to vascular injury.
Collapse
Affiliation(s)
- C H Selzman
- Department of Surgery, University of Colorado Health Sciences Center, Denver 80262, USA.
| | | | | | | | | |
Collapse
|
37
|
Selzman CH, Shames BD, Reznikov LL, Miller SA, Meng X, Barton HA, Werman A, Harken AH, Dinarello CA, Banerjee A. Liposomal delivery of purified inhibitory-kappaBalpha inhibits tumor necrosis factor-alpha-induced human vascular smooth muscle proliferation. Circ Res 1999; 84:867-75. [PMID: 10222332 DOI: 10.1161/01.res.84.8.867] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Vessel injury results in the elaboration of various cytokines, including tumor necrosis factor-alpha (TNF-alpha), which may influence vascular smooth muscle cell (VSMC) function and contribute to atherogenesis. We tested the hypothesis that TNF-alpha-induced VSMC proliferation requires activation of the transcription factor nuclear factor-kappaB (NF-kappaB), which could be prevented by delivery of the NF-kappaB inhibitory peptide, IkappaBalpha. TNF-alpha induced concentration-dependent human VSMC proliferation, and neutralizing antibody to interleukin-6 reduced TNF-alpha-induced VSMC proliferation by 65%. In TNF-alpha-stimulated VSMCs, there was a 3-fold increase in NF-kappaB-dependent luciferase reporter activity that was associated with degradation of IkappaBalpha. To determine an essential role for NF-kappaB in TNF-alpha-induced VSMC proliferation, recombinant IkappaBalpha was introduced into VSMCs via liposomal delivery. Under these conditions, TNF-alpha-induced NF-kappaB nuclear translocation and DNA binding were inhibited, NF-kappaB-dependent luciferase activity was reduced by 50%, there was no degradation of native IkappaBalpha detected, interleukin-6 production was reduced by 54%, and VSMC proliferation was decreased by 60%. In conclusion, the mitogenic effect of TNF-alpha on human arterial VSMCs is dependent on NF-kappaB activation and may be prevented by exogenously delivered IkappaBalpha. Furthermore, liposomal delivery of endogenous inhibitory proteins may represent a novel, therapeutically accessible method for selective transcriptional suppression in the response to vascular injury.
Collapse
Affiliation(s)
- C H Selzman
- Department of Surgery, University of Colorado Health Sciences Center, Denver, Colo. 80262, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Chen G, Wood EG, Wang SH, Warner TD. Expression of cyclooxygenase-2 in rat vascular smooth muscle cells is unrelated to nuclear factor-kappaB activation. Life Sci 1999; 64:1231-42. [PMID: 10210266 DOI: 10.1016/s0024-3205(99)00055-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The promoter region of cyclooxygenase-2 (COX-2) gene contains binding sites for a number of important transcription factors including cyclic AMP response element, nuclear factor-IL6, nuclear factor-kappaB (NF-kappaB) and TGF-beta response element. Several reports have documented that the activation of NF-kappaB triggers the expression of COX-2 gene. In the present study, NF-kappaB was activated by TNF-alpha in rat aortic smooth muscle cells as demonstrated by electrophoretic mobility shift assay. The activity of NF-kappaB induced by TNF-alpha was blocked by calpain inhibitor I, a potent NF-kappaB inhibitor. However, the activation of NF-kappaB was not related to the expression of COX-2 induced by TNF-alpha since calpain inhibitor I blocked the activation of NF-kappaB but not the expression of COX-2 mRNA or protein. Mutation of rat COX-2 NF-kappaB-like sites in the promoter region did not significantly reduce the promoter activity. These results suggest that the transcriptional regulation of COX-2 expression by NF-kappaB depends upon the types of cells studied and that activation of this transcription factor alone does not play an important role in the expression of COX-2 in rat smooth muscle cells.
Collapse
Affiliation(s)
- G Chen
- William Harvey Research Institute, St. Bartholomew's Hospital Medical College, Charterhouse Square, London, UK
| | | | | | | |
Collapse
|
39
|
Erl W, Hansson GK, de Martin R, Draude G, Weber KS, Weber C. Nuclear factor-kappa B regulates induction of apoptosis and inhibitor of apoptosis protein-1 expression in vascular smooth muscle cells. Circ Res 1999; 84:668-77. [PMID: 10189354 DOI: 10.1161/01.res.84.6.668] [Citation(s) in RCA: 99] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Apoptosis is important in normal development as well as in diseases such as atherosclerosis. However, the regulation of apoptosis is still not completely understood. We now show that the transcription factor nuclear factor-kappaB (NF-kappaB) controls the induction of apoptosis in human and rat vascular smooth muscle cells (SMCs). SMCs in high-density culture exhibited a high NF-kappaB activity and were insensitive to induction of apoptosis. Inhibition of NF-kappaB by adenovirus-mediated overexpression of its inhibitor IkappaBalpha caused a marked increase in cell death at low but not high cell density. Elevating endogenous IkappaBalpha levels by inhibiting its degradation with proteasomal inhibitors resulted in induction of apoptosis in low-density SMCs, as detected by increased binding of annexin V, reduced mitochondrial membrane potential, and increased hypodiploid DNA. In high-density cultures, protection against apoptosis was associated with the expression of inhibitor of apoptosis protein-1 (IAP-1). Transfer of IkappaBalpha reduced human IAP-1 mRNA levels, which suggested that IAP-1 is transcriptionally regulated by NF-kappaB. This was confirmed through identification of a motif with NF-kappaB-like binding activity in the human IAP-1 promoter region. Moreover, antisense inhibition of IAP-1 sensitized high-density SMCs to the induction of cell death. Together, our data imply that SMCs at high density are protected by an antiapoptotic mechanism that involves increased expression of NF-kappaB and IAP-1. Interference with pathways that control the susceptibility to programmed cell death may be helpful in the treatment of diseases where dysregulation of apoptosis is involved, eg, atherosclerosis and restenosis.
Collapse
Affiliation(s)
- W Erl
- Cardiovascular Research Unit, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden.
| | | | | | | | | | | |
Collapse
|
40
|
Newman WH, Zhang LM, Lee DH, Dalton ML, Warejcka DJ, Castresana MR, Leeper-Woodford SK. Release of tumor necrosis factor-alpha from coronary smooth muscle: activation of NF-kappaB and inhibition by elevated cyclic AMP. J Surg Res 1998; 80:129-35. [PMID: 9878303 DOI: 10.1006/jsre.1998.5456] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND Evidence suggests that tumor necrosis factor-alpha (TNF-alpha) is involved in heart diseases such as atherosclerosis. We used porcine coronary arteries and smooth muscle cells cultured from these vessels to study the regulation of production of TNF-alpha. The aims were to determine if bacterial lipopolysaccharide (LPS) could stimulate production; if activation of the nuclear regulatory factor, NF-kappaB, was associated with production; and if intracellular cAMP regulates TNF-alpha in coronary vasculature through a mechanism involving NF-kappaB. MATERIAL AND METHODS LPS was used to stimulate TNF-alpha production. Forskolin (FSK) and 8-Br-cAMP were added to tissue and cells in order to elevate intracellular cAMP. TNF-alpha release into the bathing medium was measured by the L929 cell cytotoxicity assay. Intracellular cAMP was determined by radioimmunoassay. NF-kappaB activation was determined in whole cell extracts by electrophoretic mobility shift assay. RESULTS In segments of coronary arteries, LPS stimulated TNF-alpha release which increased with time to a maximum at 6 h (485 +/- 19 units/g tissue) and remained elevated at this level for 24 h. In contrast, the level of TNF-alpha measured at 24 h in medium from coronary tissue not exposed to LPS was 11.1 +/- 4.1 units/g tissue. In the presence of LPS, both FSK and 8-Br-cAMP significantly reduced TNF-alpha release. For instance at 6 h in the presence of LPS and FSK or 8-Br-cAMP, TNF-alpha was 126 +/- 24 and 71.6 +/- 22 units/g tissue, respectively (P < 0.05 vs LPS alone). Tissue levels of cAMP were significantly elevated in the presence of FSK. Similar results were obtained with smooth muscle cells cultured from the coronary arteries; i.e., LPS stimulated TNF-alpha release which was inhibited in a concentration-dependent manner by a rise in intracellular cAMP induced by FSK. In cultured cells release of TNF-alpha stimulated by LPS was associated with activation of NF-kappaB. Neither FSK nor 8-Br cAMP inhibited activation of NF-kappaB by LPS. CONCLUSIONS Porcine coronary arteries produce TNF-alpha from a smooth muscle cell source. Production stimulated by LPS was inhibited by elevated intracellular cAMP and was associated with activation of NF-kappaB. However, activation of NF-kappaB was not inhibited by elevated cAMP, suggesting that the regulatory action of this cyclic nucleotide could lie downstream from activation of the TNF-alpha gene. These results support the view that coronary vessels can be a source of TNF-alpha possibly involved in heart disease.
Collapse
Affiliation(s)
- W H Newman
- Department of Surgery, Mercer University School of Medicine and Medical Center of Central Georgia, Macon, Georgia, 31201, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Lee JS, Kypreos KE, Sonenshein GE. Synchronization of cultured vascular smooth muscle cells following reversal of quiescence induced by treatment with the antioxidant N-acetylcysteine. Exp Cell Res 1998; 239:447-53. [PMID: 9521863 DOI: 10.1006/excr.1997.3919] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Smooth muscle cell (SMC) proliferation plays an important role in the pathogenesis of vascular diseases such as atherosclerosis and postangioplasty restenosis. Recently we demonstrated the thiol antioxidant N-acetylcysteine (NAC) inhibits constitutive NF-kappa B/Rel activity and growth of vascular SMCs. Here we show that treatment of human and bovine aortic SMC with the thiol antioxidant NAC causes cells to exit the cell cycle and remain quiescent as determined by a greatly reduced incorporation of [3H]thymidine and G0/G1 DNA content. Removal of NAC from the culture medium stimulates SMCs to synchronously reenter the cell cycle as judged by induction of cyclin D1 and B-myb gene expression during mid and late G1 phase, respectively, and induction of histone gene expression and [3H]thymidine incorporation during S phase. The time course of cyclin D1, B-myb, and histone gene expression after NAC removal was similar to that of serum-deprived cells induced to resume cell cycle progression by the addition of fetal bovine serum to the culture medium. Taken together, these results indicate that NAC treatment causes SMCs to enter a reversible G0 quiescent, growth-arrested state. Thus, NAC provides an important new method for synchronizing SMCs in culture.
Collapse
Affiliation(s)
- J S Lee
- Department of Biochemistry, Boston University School of Medicine, Massachusetts 02118, USA
| | | | | |
Collapse
|
42
|
Ni A, Chao L, Chao J. Transcription factor nuclear factor kappaB regulates the inducible expression of the human B1 receptor gene in inflammation. J Biol Chem 1998; 273:2784-91. [PMID: 9446586 DOI: 10.1074/jbc.273.5.2784] [Citation(s) in RCA: 115] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Expression of the bradykinin B1 receptor gene is up-regulated in vascular smooth muscle cells (VSMCs) in response to a variety of inflammatory stimuli. We isolated the 5'-flanking region of the human bradykinin B1 receptor gene and examined its promoter activity by transient transfection analysis. This region (-2582 to +34) showed promoter activity inducible by lipopolysaccharide (LPS), tumor necrosis factor alpha (TNF-alpha), and interleukin-1beta (IL-1beta) in VSMCs. Further deletion analysis revealed that constructs containing 111 base pairs of 5'-flanking sequence were sufficient for transcriptional induction. Mutagenesis of a nuclear factor kappaB (NF-kappaB)-like site at -64 to -55 abolished most of the LPS, TNF-alpha, and IL-1beta inducibility, whereas a mutation of a cyclic AMP response element at -50 to -43 markedly reduced the basal promoter activity, and a mutation of the activator protein 1 (AP-1) site at -78 to -72 had minimal effects. Nuclear extracts from LPS, TNF-alpha, and IL-1beta-treated VSMCs, IL-1beta-treated human hepatoma HepG2, and human lung fibroblast IMR-90 cells showed strong inducible binding activity to the NF-kappaB-like site by gel shift assays. These results demonstrated that NF-kappaB-like nuclear factor was involved in the inducible expression of the human bradykinin B1 receptor gene during inflammatory processes.
Collapse
MESH Headings
- Amino Acid Sequence
- Base Sequence
- Cloning, Molecular
- DNA Mutational Analysis
- Gene Expression Regulation
- Genes, Reporter
- Humans
- Inflammation
- Interleukin-1/pharmacology
- Lipopolysaccharides/pharmacology
- Molecular Sequence Data
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Mutagenesis, Site-Directed
- NF-kappa B/metabolism
- Promoter Regions, Genetic
- Protein Binding
- Receptor, Bradykinin B1
- Receptors, Bradykinin/biosynthesis
- Receptors, Bradykinin/genetics
- Tumor Necrosis Factor-alpha/pharmacology
Collapse
Affiliation(s)
- A Ni
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | |
Collapse
|
43
|
Bourcier T, Sukhova G, Libby P. The nuclear factor kappa-B signaling pathway participates in dysregulation of vascular smooth muscle cells in vitro and in human atherosclerosis. J Biol Chem 1997; 272:15817-24. [PMID: 9188479 DOI: 10.1074/jbc.272.25.15817] [Citation(s) in RCA: 197] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
In the lesions of atherosclerosis, vascular smooth muscle cells (SMC) display many functions characteristic of cytokine activation that likely contribute importantly to ongoing inflammation during human atherogenesis. The transcription factor nuclear factor kappa-B (NFkappaB) often mediates the effects of cytokines on target cells, but the identity of Rel family members important in human SMC activation remains uncertain. In vitro, human SMC express multiple Rel family members. Of these, dimers of p65 and p50, but not a putative SMC-Rel, comprise basal and inducible NFkappaB binding activities. SMC express two inhibitor proteins IkappaBbeta and IkappaBalpha. Interleukin-1beta stimulation caused transient loss of IkappaBalpha and a sustained decrease of IkappaBbeta that correlated with increased and persistent levels of p65/p50 protein and binding activity in the nucleus. SMC cultured under serum-free conditions displayed little NFkappaB activity, but addition of serum or platelet-derived growth factor did activate NFkappaB. In situ analyses showed no evidence for basal NFkappaB activity in SMC in vivo as nonatherosclerotic arteries did not contain nuclear p65 or p50 protein. However, the nuclei of intimal SMC within human atheroma did contain both Rel proteins. We conclude that (i) dimers of p65 and p50, but not SMC-Rel, comprise NFkappaB complexes in human SMC; (ii) stimulatory components in serum activate NFkappaB and likely account for previously reported "constitutive" NFkappaB activity in cultured SMC; and (iii) exposure to inflammatory cytokines may produce prolonged NFkappaB activation in SMC because of sustained decreases in the inhibitory subunit IkappaB-beta.
Collapse
Affiliation(s)
- T Bourcier
- Vascular Medicine and Atherosclerosis Unit, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
44
|
Cercek B, Yamashita M, Dimayuga P, Zhu J, Fishbein MC, Kaul S, Shah PK, Nilsson J, Regnstrom J. Nuclear factor-kappaB activity and arterial response to balloon injury. Atherosclerosis 1997; 131:59-66. [PMID: 9180245 DOI: 10.1016/s0021-9150(96)06767-6] [Citation(s) in RCA: 75] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
We studied the effect of arterial balloon injury on nuclear factor-kappaB (NF-kappaB) mobilization and ICAM-1 expression in untreated rats and rats treated with aspirin. Baseline NF-kappaB nuclear binding in smooth muscle cells (SMC) increased two-fold within 6 h after balloon injury. The binding returned to baseline 3 days after injury. Consistently nuclear staining of p65 active subunit increased in the medial SMC following balloon injury. There was no baseline ICAM-1 expression. Within 3 days after balloon injury there was marked medial ICAM-1 expression, that localized to neointima 7 days after injury and to regrowing endothelial cells 14 days after injury. Treatment with aspirin inhibited NF-kappaB nuclear translocation and binding and was associated with reduction of ICAM-1 expression, SMC proliferation and neointimal thickening following balloon injury. These data suggest that transient mobilization of NF-kappaB in vascular SMC after balloon injury mediates ICAM-1 expression and is involved in arterial response to balloon injury.
Collapse
Affiliation(s)
- B Cercek
- Atherosclerosis Research Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Schini-Kerth VB, Boese M, Busse R, Fisslthaler B, Mülsch A. N-alpha-tosyl-L-lysine chloromethylketone prevents expression of iNOS in vascular smooth muscle by blocking activation of NF-kappa B. Arterioscler Thromb Vasc Biol 1997; 17:672-9. [PMID: 9108779 DOI: 10.1161/01.atv.17.4.672] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Certain cytokines and lipopolysaccharide stimulate expression of inducible nitric oxide synthase (iNOS) in vascular smooth muscle, an event that is regulated at the transcriptional level and appears to involve several transcription factors, including nuclear factor kappa B (NF-kappa B). Since proteases play an essential role in NF-kappa B activation, experiments were designed to clarify, in both cultured rat aortic smooth muscle cells (SMCs) and isolated rat aortas, whether protease inhibitors affect the interleukin-1 beta (IL-1 beta)-elicited expression of iNOS. The formation of NO was assessed by nitrite release in cultured SMCs and the attenuation of phenylephrine-induced contraction in aortic rings, the expression of iNOS by Western blot analysis and reverse transcription-polymerase chain reaction, and NF-kappa B activity in nuclear extracts by gel electrophoretic mobility shift assya. Exposure of cultured SMCs to IL-1 beta increased NF-kappa B binding activity within 30 minutes and was associated with nitrite accumulation and the appearance of iNOS protein 24 hours later. These responses were abolished in cells that had been exposed to the cytokine in the presence of the protease inhibitor N-alpha-tosyl-L-lysine chloromethylketone. Aprotinin and p-toluenesulfonyl-L-arginine methyl ester, two other protease inhibitors, also reduced the cytokine-stimulated release of nitrite and the level of iNOS protein. Exposure of rat aortic segments without endothelium to IL-1 beta activated NF-kappa B within 30 minutes and was associated with the appearance of iNOS mRNA and an attenuation of phenylephrine-induced contraction 6 hours later. These responses were blunted when the segments were incubated with the cytokine and N-alpha-tosyl-L-lysine chloromethyl ketone. The present observations indicate that protease inhibitors prevent iNOS expression in both cultured and native vascular SMCs by blocking the activation of NF-kappa B.
Collapse
|
46
|
Renard P, Zachary MD, Bougelet C, Mirault ME, Haegeman G, Remacle J, Raes M. Effects of antioxidant enzyme modulations on interleukin-1-induced nuclear factor kappa B activation. Biochem Pharmacol 1997; 53:149-60. [PMID: 9037247 DOI: 10.1016/s0006-2952(96)00645-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Nuclear factor kappa B (NF-kappa B) is a potent and pleiotropic transcription factor that can be activated by a wide variety of inducers, including interleukin-1 (IL-1). Although the detailed activation mechanism of NF-kappa B is still under investigation, it requires both phosphorylation and degradation of its inhibitory subunit I kappa B and the presence of an oxidative environment. In this study, we systematically evaluated the influence of glutathione peroxidase, glutathione reductase and catalase on IL-1-induced NF-kappa B activation by analysing the effect of specific inhibitors of these enzymes. For the three antioxidant enzymes mentioned, their inhibition correlated with an overactivation of NF-kappa B, particularly for glutathione peroxidase. Inversely, we tested the response of glutathione peroxidase-transfected cells on NF-kappa B activation, which was lower as compared with the parental cells. Furthermore, interleukin-6 production also correlated perfectly with the reduced level of NF-kappa B activation is these experiments. The results clearly show that NF-kappa B activation is, strongly dependent on the antioxidant potential of the cells, especially on the activity of reduced glutathione-dependent enzymes such as glutathione peroxidase. The results support the hypothesis that the level of the oxidised glutathione:reduced glutathione ratio and the activity of intracellular antioxidant enzymes play a major role in NF-kappa B tine tuning.
Collapse
Affiliation(s)
- P Renard
- Laboratoire de Biochimie Cellulaire, Facultés Universitaires, Namur, Belgium
| | | | | | | | | | | | | |
Collapse
|
47
|
Clesham GJ, Browne H, Efstathiou S, Weissberg PL. Enhancer stimulation unmasks latent gene transfer after adenovirus-mediated gene delivery into human vascular smooth muscle cells. Circ Res 1996; 79:1188-95. [PMID: 8943957 DOI: 10.1161/01.res.79.6.1188] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Recombinant adenoviral vectors are being used increasingly for gene transfer studies in mammalian cells and gene therapy protocols in humans. High adenoviral titers are often required for successful transduction of vascular smooth muscle cells (VSMCs), defined as uptake and detectable expression of the foreign gene, but the relative contributions of efficiency of viral uptake and control of transcription are poorly understood. To explore the extent to which a lack of detectable gene expression may be due to inefficient transcription of a successfully transferred gene, we have used a replication-deficient adenovirus expressing beta-galactosidase (RAd35 beta-Gal), under the control of the human cytomegalovirus major immediate-early promoter (CMV-IEP), which contains cAMP and nuclear factor-kappa B response elements, to investigate constitutive and inducible gene expression after gene transfer into human VSMCs. Histochemical staining with 5-bromo-4-chloro-3-indolyl beta-D-galactopyranoside (X-gal), a quantitative spectrophotometric assay, SDS-PAGE, Western blotting, and Northern analysis were used to evaluate beta-galactosidase expression in infected cells. After infection with RAd35 beta-Gal at 30, 100, and 1000 plaque-forming units per cell (pfu/cell), expression of beta-galactosidase was augmented up to 17-, 19-, and 23-fold, respectively, in human VSMCs treated with forskolin and phorbol ester compared with unstimulated cells. After infection, the proportion of detectably transduced cells was increased by enhancer stimulation from 58% to 100% at 100 pfu/cell and from 9% to 62% at 10 pfu/cell, indicating quiescent viral DNA in unstimulated cells. At high adenoviral titers (1000 pfu/cell), the recombinant gene became the most abundant protein in cell extracts. These findings demonstrate that in human VSMCs, limited constitutive expression from the CMV-IEP, rather than failure of translocation of adenoviral DNA, may be responsible for the apparent failure of transduction at a low multiplicity of infection.
Collapse
Affiliation(s)
- G J Clesham
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, UK.
| | | | | | | |
Collapse
|
48
|
Shin WS, Hong YH, Peng HB, De Caterina R, Libby P, Liao JK. Nitric oxide attenuates vascular smooth muscle cell activation by interferon-gamma. The role of constitutive NF-kappa B activity. J Biol Chem 1996; 271:11317-24. [PMID: 8626684 DOI: 10.1074/jbc.271.19.11317] [Citation(s) in RCA: 106] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Atherogenesis involves cellular immune responses and altered vascular smooth muscle cell (SMC) function. Cytokines such as interleukin (IL)-1 alpha and interferon-gamma (IFN-gamma) may contribute to this process by activating SMC. To determine whether the anti-atherogenic mediator, nitric oxide (.NO), can modulate cytokine-induced SMC activation, we investigated the effects of various .NO-generating compounds on the expression of intercellular and vascular cell adhesion molecules (ICAM-1 and VCAM-1). Induction of ICAM-1 expression by IL-1 alpha and VCAM-1 expression by IFN-gamma was attenuated by .NO donors but not by cGMP analogues. Nuclear run-on assays and transfection studies using various VCAM-1 promoter constructs linked to the chloramphenicol acetyl-transferase reporter gene showed that .NO repressed IFN-gamma-induced VCAM-1 gene transcription, in part, through inhibition of nuclear factor-kappa B (NF-kappa B). Electrophoretic mobility shift assay revealed that SMC possess basal constitutive NF-kappa B activity, which was augmented by treatment with IL-1 alpha. In contrast, IFN-gamma induced and activated interferon regulatory factor (IRF)-1 but had little effect on basal constitutive NF-kappa B activity. .NO donors had no inhibitory effect on IRF-1 activation but did inhibit basal and IL-1 alpha-stimulated NF-kappa B activation. These findings suggest that the induction of ICAM-1 and VCAM-1 expression requires NF-kappa B activation and that .NO attenuates IFN-gamma-induced VCAM-1 expression primarily by inhibiting basal constitutive NF-kappa B activity in SMC.
Collapse
Affiliation(s)
- W S Shin
- Cardiovascular Division, Brigham & Women's Hospital, Boston, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
The transcription factor NF-kappa B has attracted widespread attention among researchers in many fields based on the following: its unusual and rapid regulation, the wide range of genes that it controls, its central role in immunological processes, the complexity of its subunits, and its apparent involvement in several diseases. A primary level of control for NF-kappa B is through interactions with an inhibitor protein called I kappa B. Recent evidence confirms the existence of multiple forms of I kappa B that appear to regulate NF-kappa B by distinct mechanisms. NF-kappa B can be activated by exposure of cells to LPS or inflammatory cytokines such as TNF or IL-1, viral infection or expression of certain viral gene products, UV irradiation, B or T cell activation, and by other physiological and nonphysiological stimuli. Activation of NF-kappa B to move into the nucleus is controlled by the targeted phosphorylation and subsequent degradation of I kappa B. Exciting new research has elaborated several important and unexpected findings that explain mechanisms involved in the activation of NF-kappa B. In the nucleus, NF-kappa B dimers bind to target DNA elements and activate transcription of genes encoding proteins involved with immune or inflammation responses and with cell growth control. Recent data provide evidence that NF-kappa B is constitutively active in several cell types, potentially playing unexpected roles in regulation of gene expression. In addition to advances in describing the mechanisms of NF-kappa B activation, excitement in NF-kappa B research has been generated by the first report of a crystal structure for one form of NF-kappa B, the first gene knockout studies for different forms of NF-kB and of I kappa B, and the implications for therapies of diseases thought to involve the inappropriate activation of NF-kappa B.
Collapse
Affiliation(s)
- A S Baldwin
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill 27599, USA
| |
Collapse
|
50
|
Miano JM, Olson EN. Expression of the smooth muscle cell calponin gene marks the early cardiac and smooth muscle cell lineages during mouse embryogenesis. J Biol Chem 1996; 271:7095-103. [PMID: 8636144 DOI: 10.1074/jbc.271.12.7095] [Citation(s) in RCA: 103] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Although several genes are considered markers for vascular smooth muscle cell (SMC) differentiation, few have been rigorously tested for SMC specificity in mammals, particularly during development where considerable overlap exists between different muscle gene programs. Here we describe the temporospatial expression pattern of the SMC calponin gene (formerly h1 or basic calponin) during mouse embryogenesis and in adult mouse tissues and cell lines. Whereas SMC calponin mRNA expression is restricted exclusively to SMCs in adult tissues, during early embryogenesis, SMC calponin transcripts are expressed throughout the developing cardiac tube as well as in differentiating SMCs. Transcription of the SMC calponin gene initiates at two closely juxtaposed sites in the absence of a consensus TATAA or initiator element. Transient transfection assays in cultured SMC demonstrated that high level SMC calponin promoter activity required no more than 549 nucleotides of 5 sequence. In contrast to the strict cell type-specificity of SMC calponin mRNA expression, the SMC calponin promoter showed activity in several cell lines that do not express the endogenous SMC calponin gene. These results demonstrate that SMC calponin responds to cardiac and smooth muscle gene regulatory programs and suggest that the cardiac and smooth muscle cell lineages may share a common gene regulatory program early in embryogenesis, which diverges as the heart matures. The finding that the isolated SMC calponin promoter is active in a wider range of cells than the endogenous SMC calponin gene also suggests that long-range repression or higher order regulatory mechanism(s) are involved in cell-specific regulation of SMC calponin expression.
Collapse
Affiliation(s)
- J M Miano
- Department of Biochemistry and Molecular Biology, University of Texas M. D. Anderson Cancer Center, Houston 77030, USA
| | | |
Collapse
|