101
|
Steib CJ, Gmelin L, Pfeiler S, Schewe J, Brand S, Göke B, Gerbes AL. Functional relevance of the cannabinoid receptor 2 - heme oxygenase pathway: a novel target for the attenuation of portal hypertension. Life Sci 2013; 93:543-51. [PMID: 24007798 DOI: 10.1016/j.lfs.2013.08.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 08/13/2013] [Accepted: 08/20/2013] [Indexed: 12/17/2022]
Abstract
AIMS In liver cirrhosis, inflammation triggers portal hypertension. Kupffer cells (KC) produce vasoconstrictors upon activation by bacterial constituents. Here, we hypothesize that the anti-inflammatory action of the cannabinoid receptor 2 (CB2) agonists JWH-133 and GP 1a attenuate portal hypertension. MAIN METHODS In vivo measurements of portal pressures and non-recirculating liver perfusions were performed in rats 4weeks after bile duct ligation (BDL). Zymosan (150μg/ml, isolated liver perfusion) or LPS (4mg/kgb.w., in vivo) was infused to activate the KC in the absence or presence of JWH-133 (10mg/kgb.w.), GP 1a (2.5mg/kgb.w.) or ZnPP IX (1μM). Isolated KC were treated with Zymosan (0.5mg/ml) in addition to JWH-133 (5μM). The thromboxane (TX) B2 levels in the perfusate and KC media were determined by ELISA. Heme oxygenase-1 (HO-1) and CB2 were analyzed by Western blot or confocal microscopy. KEY FINDINGS JWH-133 or GP 1a pre-treatment attenuated portal pressures following KC activation in all experimental settings. In parallel, HO-1 expression increased with JWH-133 pre-treatment. However, the inhibition of HO-1 enhanced portal hypertension, indicating the functional role of this novel pathway. In isolated KC, the expression of CB2 and HO-1 increased with Zymosan, LPS and JWH-133 treatment while TXB2 production following KC activation was attenuated by JWH-133 pre-treatment. SIGNIFICANCE JWH-133 or GP 1a treatment attenuates portal hypertension. HO-1 induction by JWH-133 plays a functional role. Therefore, the administration of JWH-133 or GP 1a represents a promising new treatment option for portal hypertension triggered by microbiological products.
Collapse
Key Words
- (6aR,10aR)-3-(1,1-dimethylbutyl)-6a,7,10,10a-tetrahydro-6,6,9-trimethyl-6H-dibenzo[b,d]pyran
- 2-Chloro-5-nitro-N-phenylbenzamide
- BDL
- Bile duct ligation (BDL)
- CB(2)
- GW 9662
- Gp 1a
- HO
- Heme oxygenase (HO)
- JWH-133
- KC
- Kupffer cell
- LDH
- LPS
- N-(Piperidin-1-yl)-1-(2,4-dichlorophenyl)-1,4-dihydro-6-methylindeno[1,2-c]pyrazole-3-carboxamide
- TLR
- TX
- Thromboxane
- Zy
- Zymosan A
- b. w
- bile duct ligation
- body weight
- cannabinoid receptor 2
- heme oxygenase
- lactate dehydrogenase
- lipopolysaccharide
- thromboxane
- toll like receptor
Collapse
Affiliation(s)
- Christian J Steib
- Department of Medicine II (Gastroenterology and Hepatology), Liver Center Munich, University of Munich, Grosshadern, Munich, Germany.
| | | | | | | | | | | | | |
Collapse
|
102
|
McCarthy FP, Delany AC, Kenny LC, Walsh SK. PPAR-γ -- a possible drug target for complicated pregnancies. Br J Pharmacol 2013. [PMID: 23186152 DOI: 10.1111/bph.12069] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Peroxisome proliferator activated receptors (PPARs) are ligand-activated transcription factors expressed in trophoblasts, which regulate both cell differentiation and proliferation. In recent years, evidence has linked PPARs to playing an integral role in pregnancy; specifically, PPAR-β and PPAR-γ have been shown to play an integral role in placentation, with PPAR-γ additionally serving to regulate trophoblast differentiation. Recent evidence has shown that PPAR-γ expression is altered in many complications of pregnancy such as intrauterine growth restriction (IUGR), preterm birth, pre-clampsia and gestational diabetes. Thus, at present, accumulating evidence from the literature suggests both a pivotal role for PPAR-γ in the progression of a healthy pregnancy and the possibility that PPAR-γ may act as a therapeutic target in complicated pregnancies. This review aims to provide a succinct and comprehensive assessment of the role of PPAR-γ in normal pregnancy and pregnancy complications, and finally its potential as a therapeutic target in the treatment and/or prevention of adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Fergus P McCarthy
- Department of Obstetrics & Gynaecology, Anu Research Centre, University College Cork, Cork, Ireland
| | | | | | | |
Collapse
|
103
|
Barlaka E, Ledvényiová V, Galatou E, Ferko M, Čarnická S, Ravingerová T, Lazou A. Delayed cardioprotective effects of WY-14643 are associated with inhibition of MMP-2 and modulation of Bcl-2 family proteins through PPAR-α activation in rat hearts subjected to global ischaemia–reperfusion. Can J Physiol Pharmacol 2013; 91:608-16. [DOI: 10.1139/cjpp-2012-0412] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors regulating cardiac lipid metabolism and energy homeostasis. Although the activation of PPARs has been implicated in cardioprotection, the molecular mechanisms are largely unexplored. In this study, we aimed to investigate the effect of the PPAR-α agonist WY-14643 (WY), mimicking a delayed effect of preconditioning in rat hearts exposed to acute ischaemia–reperfusion (I/R) 24 h later, and to define whether antioxidative and antiapoptotic mechanisms are involved. Treatment with WY markedly attenuated post-ischaemic contractile dysfunction (as evidenced by the reduced infarct size), the higher left ventricular developed pressure (LVDP) recovery, and the decreased occurrence of arrhythmias. These effects were abolished in the presence of the PPAR-α antagonist MK886. Heme oxygenase-1, a key antioxidative enzyme implicated in cytoprotection, was upregulated in response to WY at baseline, but was markedly reduced after I/R, indicating reduced oxidative stress. WY treatment was also associated with decreased mRNA levels and enzymatic activity of matrix metalloproteinase-2, and increased ratios of Bcl-2:Bax proteins. These results indicate that PPAR-α activation by its selective ligand WY may confer delayed preconditioning-like protection in rat hearts subjected to I/R by modulating oxidative stress, activation of matrix metalloproteinase-2, and expression of Bcl-2 and Bax.
Collapse
Affiliation(s)
- Eleftheria Barlaka
- School of Biology, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Veronika Ledvényiová
- Institute for Heart Research, Slovak Academy of Sciences and Centre of Excellence of SAS NOREG, Bratislava, Slovak Republic
| | - Eleftheria Galatou
- School of Biology, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Miroslav Ferko
- Institute for Heart Research, Slovak Academy of Sciences and Centre of Excellence of SAS NOREG, Bratislava, Slovak Republic
| | - Slávka Čarnická
- Institute for Heart Research, Slovak Academy of Sciences and Centre of Excellence of SAS NOREG, Bratislava, Slovak Republic
| | - Táňa Ravingerová
- Institute for Heart Research, Slovak Academy of Sciences and Centre of Excellence of SAS NOREG, Bratislava, Slovak Republic
| | - Antigone Lazou
- School of Biology, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| |
Collapse
|
104
|
Xue J, Wang S, Wu J, Hannafon BN, Ding WQ. Zinc at sub-cytotoxic concentrations induces heme oxygenase-1 expression in human cancer cells. Cell Physiol Biochem 2013; 32:100-10. [PMID: 23868099 DOI: 10.1159/000350128] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2013] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND/AIMS This study investigated the effects of zinc on heme oxygenase-1 (HO-1) expression in human cancer cells. METHODS/RESULTS Zinc at sub-cytotoxic concentrations (50-100 μM) induces HO-1 expression in the MDA-MB-231 (human breast cancer) and A2780 (human ovarian cancer) cell lines in a concentration- and time-dependent manner. The induction of HO-1 by zinc was detected after 4-6 hours of treatment, reached maximal level at 8 hours, and declined thereafter. Using a human HO-1 gene promoter reporter construct, we identified two antioxidant response elements (AREs) that mediated the zinc-induced increase in HO-1 gene transcription, indicating that the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) signaling pathway is involved in this event. This assumption was supported by the observations that knockdown of Nrf2 expression compromised the zinc-induced increase in HO-1 gene transcription, and that zinc increased Nrf2 protein expression and the Nrf2 binding to the AREs. Additionally, we found that the zinc-induced HO-1 gene transcription can be enhanced by clioquinol, a zinc ionophore, and reversed by pretreatment with TPEN, a known zinc chelator, indicating that an increase in intracellular zinc levels is responsible for this induction. CONCLUSION These findings demonstrate that zinc at sub-cytotoxic concentrations induces HO-1 expression in human cancer cells. The biological significance of this induction merits further investigation.
Collapse
Affiliation(s)
- Jing Xue
- School of Radiation Medicine and Protection, Soochow University, Suzhou, P. R. China
| | | | | | | | | |
Collapse
|
105
|
Pioglitazone ameliorates systolic and diastolic cardiac dysfunction in rat model of angiotensin II-induced hypertension. Int J Cardiol 2013; 167:409-15. [DOI: 10.1016/j.ijcard.2012.01.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 10/16/2011] [Accepted: 01/01/2012] [Indexed: 12/31/2022]
|
106
|
Guenzl PM, Raim R, Kral J, Brunner J, Sahin E, Schabbauer G. Insulin hypersensitivity induced by hepatic PTEN gene ablation protects from murine endotoxemia. PLoS One 2013; 8:e67013. [PMID: 23825606 PMCID: PMC3692528 DOI: 10.1371/journal.pone.0067013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 05/14/2013] [Indexed: 01/04/2023] Open
Abstract
Sepsis still remains a major cause for morbidity and mortality in patients. The molecular mechanisms underlying the disease are still enigmatic. A great number of therapeutic approaches have failed and treatment strategies are limited to date. Among those few admitted for clinical intervention, intensive insulin treatment has proven to be effective in the reduction of disease related complications in critically ill patients. Insulin effectively reduces glucose levels and thereby contributes to protection. On the other hand insulin is a potent signaling pathway activator. One of those is the PI3K signaling axis. Activation of PI3K is known to limit pro-inflammatory gene expression. Here we can show that in a mouse model of insulin hypersensitivity induced by the deletion of the PI3K antagonist PTEN, specifically in hepatic tissue, significant protection is conferred in murine models of lethal endotoxemia and sepsis. Acute inflammatory responses are diminished, glucose metabolism normalized and vascular activation is reduced. Furthermore we investigated the hepatic gene expression profile of relevant anti-inflammatory genes in PTEN deficient mice and found marked upregulation of PPARγ and HO-1. We conclude from our data that insulin hypersensitivity via sustained activation of the PI3K signaling pathway exerts protective effects in acute inflammatory processes.
Collapse
Affiliation(s)
- Philipp M. Guenzl
- Institute for Physiology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Roman Raim
- Institute for Physiology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Julia Kral
- Institute for Physiology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Julia Brunner
- Institute for Physiology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Emine Sahin
- Institute for Physiology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Gernot Schabbauer
- Institute for Physiology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- * E-mail:
| |
Collapse
|
107
|
Peroxisome proliferator-activated receptor-gamma agonist rosiglitazone attenuates inflammatory pain through the induction of heme oxygenase-1 in macrophages. Pain 2013; 154:1402-12. [PMID: 23707273 DOI: 10.1016/j.pain.2013.04.039] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 04/10/2013] [Accepted: 04/17/2013] [Indexed: 11/23/2022]
Abstract
Macrophage infiltration to inflammatory sites promotes tissue repair and may be involved in pain hypersensitivity. Peroxisome proliferator-activated receptor (PPAR)γ signaling is known to regulate polarity of macrophages, which are often referred to as proinflammatory (M1) and antiinflammatory (M2) macrophages. We recently showed that the PPARγ agonist rosiglitazone ameliorated the development of postincisional hyperalgesia by increasing the influx of M2 macrophages to inflamed sites. It has been suggested that heme oxygenase (HO)-1, upregulated by PPARγ signaling, promotes differentiation of macrophages to M2 phenotype. In this study, we investigated how rosiglitazone alters pain hypersensitivity by a PPARγHO-1-dependent mechanism during the course of inflammation induced by complete Freund's adjuvant. Local administration of rosiglitazone alleviated mechanical hyperalgesia, with increased gene induction of HO-1. Phenotype switching of infiltrated macrophages to M2 by rosiglitazone was reversed by an HO-1 inhibitor, tin protoporphyrin, at the inflamed sites. Direct stimulation of peritoneal macrophages with rosiglitazone also increased HO-1 induction in the presence of lipopolysaccharide/interferon-γ. Moreover, rosiglitazone increased gene induction of endogenous opioid proenkephalin, both at inflamed sites and in isolated macrophages. Administration of naloxone blocked the analgesic effects of rosiglitazone. We speculate that rosiglitazone alleviated the development of inflammatory pain, possibly through regulating the M1/M2 balance at the inflamed site by a PPARγ/HO-1-dependent mechanism. PPARγ signaling in macrophages may be a potential therapeutic target for the treatment of acute pain development.
Collapse
|
108
|
Levytska K, Kingdom J, Baczyk D, Drewlo S. Heme oxygenase-1 in placental development and pathology. Placenta 2013; 34:291-8. [DOI: 10.1016/j.placenta.2013.01.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 01/08/2013] [Accepted: 01/12/2013] [Indexed: 12/30/2022]
|
109
|
Hu YF, Lee KT, Wang HH, Ueng KC, Yeh HI, Chao TF, Liao JN, Lin YJ, Chang SL, Lo LW, Tuan TC, Li CH, Chung FP, Hsu CP, Chang HH, Huang CH, Chen SA. The association between heme oxygenase-1 gene promoter polymorphism and the outcomes of catheter ablation of atrial fibrillation. PLoS One 2013; 8:e56440. [PMID: 23437133 PMCID: PMC3577889 DOI: 10.1371/journal.pone.0056440] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 01/08/2013] [Indexed: 01/26/2023] Open
Abstract
A length polymorphism of GT repeats in the promoter region of the human heme oxygenase-1 (HO-1) gene modulates its gene transcription to protect against myocardial injury. The present study investigated the association between HO-1 promoter polymorphisms and the outcomes of catheter ablation of atrial fibrillation (AF). The allelic frequencies of GT repeats in the HO-1 gene promoter were screened in 205 random individuals who underwent catheter ablation for drug refractory AF.In the patients who received catheter ablation, those with AF recurrence had fewer GT repeats (53.4±7.1 vs. 56.1±6.5, p = 0.004), a lower incidence of hyperlipidemia, more non-paroxysmal AF, and a larger left atrial diameter. After conducting a multivariate logistic analysis, the number of GT repeats (Odds ratio: 0.94, 95% CI 0.90–0.99, p = 0.01) and the diameter of the left atrium (Odds ratio: 1.08, 95% CI 1.02–1.15, p = 0.01) remained independent predictors. The carriers of GT repeats, which were <29 in both alleles, were associated with a lower sinus maintenance rate after catheter ablation (38.5% vs. 60.1%, p = 0.003). The patients were divided into paroxysmal and non-paroxysmal AF groups; the number of GT repeats was associated with AF recurrence only in the patients with paroxysmal AF. The number of GT repeats, combined with LAD, was significant for predicting AF recurrence after catheter ablation (p = 0.01). The number of GT repeats was not found to be associated with differences in the left atrial diameter, the biatrial voltage, or the levels of bilirubin, ferritin, iron, C-reactive protein, or von-Willibrand factor. In conclusions, HO-1 gene promoter polymorphisms were associated with AF recurrence after catheter ablation.
Collapse
Affiliation(s)
- Yu-Feng Hu
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and Cardiovascular Research Institute, National Yang-Ming University, Taipei, Taiwan
| | - Kun-Tai Lee
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Hsueh-Hsiao Wang
- Departments of Internal Medicine and Medical Research, Mackay Memorial Hospital, Mackay Medical College, Taipei, Taiwan
| | - Kwo-Chang Ueng
- Chung Shan Medical University Hospital, Department of Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Hung-I Yeh
- Departments of Internal Medicine and Medical Research, Mackay Memorial Hospital, Mackay Medical College, Taipei, Taiwan
| | - Tze-Fan Chao
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and Cardiovascular Research Institute, National Yang-Ming University, Taipei, Taiwan
| | - Jo-Nan Liao
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yenn-Jiang Lin
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and Cardiovascular Research Institute, National Yang-Ming University, Taipei, Taiwan
| | - Shih-Lin Chang
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and Cardiovascular Research Institute, National Yang-Ming University, Taipei, Taiwan
| | - Li-Wei Lo
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and Cardiovascular Research Institute, National Yang-Ming University, Taipei, Taiwan
| | - Ta-Chuan Tuan
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and Cardiovascular Research Institute, National Yang-Ming University, Taipei, Taiwan
| | - Cheng-Hung Li
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and Cardiovascular Research Institute, National Yang-Ming University, Taipei, Taiwan
| | - Fa-Po Chung
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and Cardiovascular Research Institute, National Yang-Ming University, Taipei, Taiwan
| | - Chiao-Po Hsu
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, and National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Hsiao-Huang Chang
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, and National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Cheng-Hsiung Huang
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, and National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Shih-Ann Chen
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and Cardiovascular Research Institute, National Yang-Ming University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
110
|
Short-chain fatty acids stimulate angiopoietin-like 4 synthesis in human colon adenocarcinoma cells by activating peroxisome proliferator-activated receptor γ. Mol Cell Biol 2013; 33:1303-16. [PMID: 23339868 DOI: 10.1128/mcb.00858-12] [Citation(s) in RCA: 228] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Angiopoietin-like protein 4 (ANGPTL4/FIAF) has been proposed as a circulating mediator between the gut microbiota and fat storage. Here, we show that transcription and secretion of ANGPTL4 in human T84 and HT29 colon adenocarcinoma cells is highly induced by physiological concentrations of short-chain fatty acids (SCFA). SCFA induce ANGPTL4 by activating the nuclear receptor peroxisome proliferator activated receptor γ (PPARγ), as demonstrated using PPARγ antagonist, PPARγ knockdown, and transactivation assays, which show activation of PPARγ but not PPARα and PPARδ by SCFA. At concentrations required for PPARγ activation and ANGPTL4 induction in colon adenocarcinoma cells, SCFA do not stimulate PPARγ in mouse 3T3-L1 and human SGBS adipocytes, suggesting that SCFA act as selective PPARγ modulators (SPPARM), which is supported by coactivator peptide recruitment assay and structural modeling. Consistent with the notion that fermentation leads to PPAR activation in vivo, feeding mice a diet rich in inulin induced PPAR target genes and pathways in the colon. We conclude that (i) SCFA potently stimulate ANGPTL4 synthesis in human colon adenocarcinoma cells and (ii) SCFA transactivate and bind to PPARγ. Our data point to activation of PPARs as a novel mechanism of gene regulation by SCFA in the colon, in addition to other mechanisms of action of SCFA.
Collapse
|
111
|
Haskew-Layton RE, Payappilly JB, Xu H, Bennett SAL, Ratan RR. 15-Deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) protects neurons from oxidative death via an Nrf2 astrocyte-specific mechanism independent of PPARγ. J Neurochem 2013. [DOI: 10.1111/jnc.12107] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Renée E. Haskew-Layton
- The Burke Medical Research Institute; Department of Neurology and Neuroscience; Weill Medical College of Cornell University; White Plains New York USA
| | - Jimmy B. Payappilly
- The Burke Medical Research Institute; Department of Neurology and Neuroscience; Weill Medical College of Cornell University; White Plains New York USA
| | - Hongbin Xu
- Neural Regeneration Laboratory and Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology, and Immunology; University of Ottawa; Ottawa Canada
| | - Steffany A. L. Bennett
- Neural Regeneration Laboratory and Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology, and Immunology; University of Ottawa; Ottawa Canada
| | - Rajiv R. Ratan
- The Burke Medical Research Institute; Department of Neurology and Neuroscience; Weill Medical College of Cornell University; White Plains New York USA
| |
Collapse
|
112
|
Kim DH, Liu J, Bhat S, Benedict G, Lecka-Czernik B, Peterson SJ, Ebraheim NA, Heck BE. Peroxisome proliferator-activated receptor delta agonist attenuates nicotine suppression effect on human mesenchymal stem cell-derived osteogenesis and involves increased expression of heme oxygenase-1. J Bone Miner Metab 2013; 31:44-52. [PMID: 22945906 DOI: 10.1007/s00774-012-0382-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 07/25/2012] [Indexed: 12/24/2022]
Abstract
Smoking has long been associated with osteoporosis, decreased bone mineral density, increased risk of bone fracture, and increased health costs. Nicotine, the main component of cigarette smoke, has major negative effects on bone metabolism and skeletal remodeling in vivo. Although osteoblasts and osteoblast-like cells have been used extensively to study the impact of nicotine, few studies have been performed on human mesenchymal stem cells (hMSCs). In this context, we examined the impact of nicotine on (a) hMSCs proliferation, (b) osteoblastic differentiation, (c) alkaline phosphatase (ALP) activity, and (d) expression of canonical genes during differentiation of hMSCs. MSCs isolated from human bone marrow were treated with different concentrations (0, 0.1, 1 and 10 μM) of nicotine for 7 days. Nicotine caused a dose-dependent decrease in cell proliferation, decreased heme oxygenase-1 (HO-1) expression (p < 0.05) and attenuated osteogenesis (p < 0.05) in hMSCs (45 % reduction at day 14). In addition, nicotine caused a dose-dependent decrease in alizarin red staining for calcium and staining for ALP. Induction of HO-1 by peroxisome proliferator-activated receptor delta agonist (GW0742) prevented the effect of nicotine. Nicotine caused a dose-dependent reduction in the expression of BMP-2, a well-known marker for bone formation; however, this was prevented by GW0742 treatment. Therefore, induction of HO-1 prevents the deleterious effects of nicotine on osteogenesis in hMSC. This offers insight into both how nicotine affects bone remodeling and a therapeutic approach to prevent fracture and osteoporosis in smokers.
Collapse
Affiliation(s)
- Dong Hyun Kim
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA.
| | | | | | | | | | | | | | | |
Collapse
|
113
|
Eirin A, Li Z, Zhang X, Krier JD, Woollard JR, Zhu XY, Tang H, Herrmann SM, Lerman A, Textor SC, Lerman LO. A mitochondrial permeability transition pore inhibitor improves renal outcomes after revascularization in experimental atherosclerotic renal artery stenosis. Hypertension 2012; 60:1242-9. [PMID: 23045468 DOI: 10.1161/hypertensionaha.112.199919] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Revascularization improves blood pressure but not renal function in most patients with atherosclerotic renal artery stenosis (ARAS), possibly related to injury incurred during renal reperfusion. Bendavia, a novel tetrapeptide that inhibits mitochondrial permeability transition pore opening, reduces apoptosis, oxidative stress, and ischemia-reperfusion injury in experimental models. However, its potential for improving renal response to revascularization of chronic ARAS is unknown. We hypothesized that adjunct Bendavia would improve renal structure and function after percutaneous transluminal renal angioplasty (PTRA). Pigs were treated after 6 weeks of ARAS or control with PTRA+stenting (or sham), adjunct continuous 4-hour infusion of Bendavia (0.05 mg/kg IV) or vehicle (n=7 each) during PTRA. Single-kidney renal blood flow and glomerular filtration rate were studied 4 weeks later and renal mitochondrial biogenesis, microvascular architecture, and injurious pathways evaluated ex vivo. Monocyte chemoattractant protein-1 levels rose after PTRA, suggesting inflammatory injury. Bendavia did not immediately affect inflammatory cytokine levels, yet 4 weeks later, stenotic kidney renal blood flow and glomerular filtration rate both improved (44.00 ± 0.21% and 36.40 ± 10.21%, respectively) in ARAS+PTRA+Bendavia compared with ARAS+PTRA+vehicle. Renal mitochondrial biogenesis was restored after PTRA+Bendavia, and microvascular rarefaction, apoptosis, oxidative stress, tubular injury, and fibrosis decreased. Infusion of Bendavia during PTRA preserved mitochondrial biogenesis, renal hemodynamics, and function, and attenuated tissue injury in swine ARAS. Thus, functional mitochondrial injury during renal reperfusion may sustain renal inflammatory injury and limit kidney recovery after PTRA. Potent antiapoptotic and antioxidant effects provide Bendavia a novel therapeutic potential for improving kidney outcomes after PTRA in experimental ARAS.
Collapse
Affiliation(s)
- Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
114
|
Regulation of ROS production and vascular function by carbon monoxide. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2012; 2012:794237. [PMID: 22928087 PMCID: PMC3425856 DOI: 10.1155/2012/794237] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 07/04/2012] [Indexed: 01/06/2023]
Abstract
Carbon monoxide (CO) is a gaseous molecule produced from heme by heme oxygenase (HO). CO interacts with reduced iron of heme-containing proteins, leading to its involvement in various cellular events via its production of mitochondrial reactive oxygen species (ROS). CO-mediated ROS production initiates intracellular signal events, which regulate the expression of adaptive genes implicated in oxidative stress and functions as signaling molecule for promoting vascular functions, including angiogenesis and mitochondrial biogenesis. Therefore, CO generated either by exogenous delivery or by HO activity can be fundamentally involved in regulating mitochondria-mediated redox cascades for adaptive gene expression and improving blood circulation (i.e., O2 delivery) via neovascularization, leading to the regulation of mitochondrial energy metabolism. This paper will highlight the biological effects of CO on ROS generation and cellular redox changes involved in mitochondrial metabolism and angiogenesis. Moreover, cellular mechanisms by which CO is exploited for disease prevention and therapeutic applications will also be discussed.
Collapse
|
115
|
Schmidt S, Stahl F, Mutz KO, Scheper T, Hahn A, Schuchardt JP. Transcriptome-based identification of antioxidative gene expression after fish oil supplementation in normo- and dyslipidemic men. Nutr Metab (Lond) 2012; 9:45. [PMID: 22621246 PMCID: PMC3408332 DOI: 10.1186/1743-7075-9-45] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 05/23/2012] [Indexed: 12/31/2022] Open
Abstract
Background The beneficial effects of omega-3 polyunsaturated fatty acids (n-3 PUFAs), especially in dyslipidemic subjects with a high risk of cardiovascular disease, are widely described in the literature. A lot of effects of n-3 PUFAs and their oxidized metabolites are triggered by regulating the expression of genes. Currently, it is uncertain if the administration of n-3 PUFAs results in different expression changes of genes related to antioxidative mechanisms in normo- and dyslipidemic subjects, which may partly explain their cardioprotective effects. The aim of this study was to investigate the effects of n-3 PUFA supplementation on expression changes of genes involved in oxidative processes. Methods Ten normo- and ten dyslipidemic men were supplemented for twelve weeks with fish oil capsules, providing 1.14 g docosahexaenoic acid and 1.56 g eicosapentaenoic acid. Gene expression levels were determined by whole genome microarray analysis and quantitative real-time polymerase chain reaction (qRT-PCR). Results Using microarrays, we discovered an increased expression of antioxidative enzymes and a decreased expression of pro-oxidative and tissue enzymes, such as cytochrome P450 enzymes and matrix metalloproteinases, in both normo- and dyslipidemic men. An up-regulation of catalase and heme oxigenase 2 in both normo- and dyslipidemic subjects and an up-regulation of cytochrome P450 enzyme 1A2 only in dyslipidemic subjects could be observed by qRT-PCR analysis. Conclusions Supplementation of normo- and dyslipidemic subjects with n-3 PUFAs changed the expression of genes related to oxidative processes, which may suggest antioxidative and potential cardioprotective effects of n-3 PUFAs. Further studies combining genetic and metabolic endpoints are needed to verify the regulative effects of n-3 PUFAs in antioxidative gene expression to better understand their beneficial effects in health and disease prevention. Trial registration ClinicalTrials.gov (ID: NCT01089231)
Collapse
Affiliation(s)
- Simone Schmidt
- Faculty of Natural Sciences at the Leibniz University of Hannover, Institute of Food Science and Human Nutrition, Am Kleinen Felde 30, 30167, Hannover, Germany
| | - Frank Stahl
- Faculty of Natural Sciences at the Leibniz University of Hannover, Institute of Technical Chemistry, Callinstr 5, 30167, Hannover, Germany
| | - Kai-Oliver Mutz
- Faculty of Natural Sciences at the Leibniz University of Hannover, Institute of Technical Chemistry, Callinstr 5, 30167, Hannover, Germany
| | - Thomas Scheper
- Faculty of Natural Sciences at the Leibniz University of Hannover, Institute of Technical Chemistry, Callinstr 5, 30167, Hannover, Germany
| | - Andreas Hahn
- Faculty of Natural Sciences at the Leibniz University of Hannover, Institute of Food Science and Human Nutrition, Am Kleinen Felde 30, 30167, Hannover, Germany
| | - Jan Philipp Schuchardt
- Faculty of Natural Sciences at the Leibniz University of Hannover, Institute of Food Science and Human Nutrition, Am Kleinen Felde 30, 30167, Hannover, Germany
| |
Collapse
|
116
|
Luz NF, Andrade BB, Feijó DF, Araújo-Santos T, Carvalho GQ, Andrade D, Abánades DR, Melo EV, Silva AM, Brodskyn CI, Barral-Netto M, Barral A, Soares RP, Almeida RP, Bozza MT, Borges VM. Heme oxygenase-1 promotes the persistence of Leishmania chagasi infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2012; 188:4460-7. [PMID: 22461696 PMCID: PMC3331931 DOI: 10.4049/jimmunol.1103072] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Visceral leishmaniasis (VL) remains a major public health problem worldwide. This disease is highly associated with chronic inflammation and a lack of the cellular immune responses against Leishmania. It is important to identify major factors driving the successful establishment of the Leishmania infection to develop better tools for the disease control. Heme oxygenase-1 (HO-1) is a key enzyme triggered by cellular stress, and its role in VL has not been investigated. In this study, we evaluated the role of HO-1 in the infection by Leishmania infantum chagasi, the causative agent of VL cases in Brazil. We found that L. chagasi infection or lipophosphoglycan isolated from promastigotes triggered HO-1 production by murine macrophages. Interestingly, cobalt protoporphyrin IX, an HO-1 inductor, increased the parasite burden in both mouse and human-derived macrophages. Upon L. chagasi infection, macrophages from Hmox1 knockout mice presented significantly lower parasite loads when compared with those from wild-type mice. Furthermore, upregulation of HO-1 by cobalt protoporphyrin IX diminished the production of TNF-α and reactive oxygen species by infected murine macrophages and increased Cu/Zn superoxide dismutase expression in human monocytes. Finally, patients with VL presented higher systemic concentrations of HO-1 than healthy individuals, and this increase of HO-1 was reduced after antileishmanial treatment, suggesting that HO-1 is associated with disease susceptibility. Our data argue that HO-1 has a critical role in the L. chagasi infection and is strongly associated with the inflammatory imbalance during VL. Manipulation of HO-1 pathways during VL could serve as an adjunctive therapeutic approach.
Collapse
MESH Headings
- Animals
- Brazil
- Gene Expression Regulation, Enzymologic/drug effects
- Gene Expression Regulation, Enzymologic/genetics
- Gene Expression Regulation, Enzymologic/immunology
- Glycosphingolipids/immunology
- Glycosphingolipids/metabolism
- Glycosphingolipids/pharmacology
- Heme Oxygenase-1/biosynthesis
- Heme Oxygenase-1/genetics
- Heme Oxygenase-1/immunology
- Humans
- Leishmania/immunology
- Leishmania/metabolism
- Leishmaniasis, Visceral/enzymology
- Leishmaniasis, Visceral/genetics
- Leishmaniasis, Visceral/immunology
- Leishmaniasis, Visceral/pathology
- Macrophages, Peritoneal/enzymology
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/parasitology
- Macrophages, Peritoneal/pathology
- Membrane Proteins/biosynthesis
- Membrane Proteins/genetics
- Membrane Proteins/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Mice, SCID
- Photosensitizing Agents/pharmacology
- Protoporphyrins/pharmacology
- Up-Regulation/drug effects
- Up-Regulation/genetics
- Up-Regulation/immunology
Collapse
Affiliation(s)
- Nívea F Luz
- Centro de Pesquisas Gonçalo Moniz/Fundação Oswaldo Cruz, Salvador 40295-001, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
117
|
Sakamoto A, Hongo M, Saito K, Nagai R, Ishizaka N. Reduction of renal lipid content and proteinuria by a PPAR-γ agonist in a rat model of angiotensin II-induced hypertension. Eur J Pharmacol 2012; 682:131-6. [DOI: 10.1016/j.ejphar.2012.02.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 02/09/2012] [Accepted: 02/15/2012] [Indexed: 10/28/2022]
|
118
|
Yang H, Zhao LF, Zhao ZF, Wang Y, Zhao JJ, Zhang L. Heme oxygenase-1 prevents liver fibrosis in rats by regulating the expression of PPARγ and NF-κB. World J Gastroenterol 2012; 18:1680-8. [PMID: 22529699 PMCID: PMC3325536 DOI: 10.3748/wjg.v18.i14.1680] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 10/17/2011] [Accepted: 01/22/2012] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effects of heme oxygenase (HO)-1 on liver fibrosis and the expression of peroxisome proliferator-activated receptor gamma (PPARγ) and nuclear factor-kappa B (NF-κB) in rats.
METHODS: Sixty Wistar rats were used to construct liver fibrosis models and were randomly divided into 5 groups: group A (normal, untreated), group B (model for 4 wk, untreated), group C (model for 6 wk, untreated), group D [model for 6 wk, treated with zinc protoporphyrin IX (ZnPP-IX) from week 4 to week 6], group E (model for 6 wk, treated with hemin from week 4 to week 6). Next, liver injury was assessed by measuring serum alanine aminotransferase (ALT), aspartate aminotransferase (AST) and albumin levels. The degree of hepatic fibrosis was evaluated by measuring serum hyaluronate acid (HA), type IV collagen (IV-C) and by histological examination. Hydroxyproline (Hyp) content in the liver homogenate was determined. The expression levels of alpha-smooth muscle actin (α-SMA) in liver tissue were measured by real-time quantitative polymerase chain reaction (RT-PCR). The expression levels of PPARγ and NF-κB were determined by RT-PCR and Western blotting.
RESULTS: The expression of HO-1 increased with the development of fibrosis. Induction of HO-1 by hemin significantly attenuated the severity of liver injury and the levels of liver fibrosis as compared with inhibition of HO-1 by ZnPP-IX. The concentrations of serum ALT, AST, HA and IV-C in group E decreased compared with group C and group D (P < 0.01). Amount of Hyp and α-SMA in the liver tissues in group E decreased compared with group C (0.62 ± 0.14 vs 0.84 ± 0.07, 1.42 ± 0.17 vs 1.84 ± 0.17, respectively, P < 0.01) and group D (0.62 ± 0.14 vs 1.11 ± 0.16, 1.42 ± 0.17 vs 2.56 ± 0.37, respectively, P < 0.01). The expression of PPARγ at levels of transcription and translation decreased with the development of fibrosis especially in group D; and it increased in group E compared with groups C and D (0.88 ± 0.15 vs 0.56 ± 0.19, 0.88 ± 0.15 vs 0.41 ± 0.11, respectively, P < 0.01). The expression of NF-κB increased with the development of fibrosis especially in group D; and it decreased in group E compared with groups C and D (1.43 ± 0.31 vs 1.89 ± 0.29, 1.43 ± 0.31 vs 2.53 ± 0.54, respectively, P < 0.01).
CONCLUSION: Our data demonstrate a potential mechanism that HO-1 can prevent liver fibrosis by enhancing the expression of PPARγ and decreasing the expression of NF-κB in liver tissues.
Collapse
|
119
|
Shih PH, Hwang SL, Yeh CT, Yen GC. Synergistic effect of cyanidin and PPAR agonist against nonalcoholic steatohepatitis-mediated oxidative stress-induced cytotoxicity through MAPK and Nrf2 transduction pathways. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2012; 60:2924-2933. [PMID: 22364184 DOI: 10.1021/jf300005v] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) is caused by an elevation in oxidative stress, which might further lead to hepatic fibrogenesis. Importantly, both peroxisome proliferator-activated receptor (PPAR) and nuclear factor erythroid 2-related factor 2 (Nrf2) play roles in modulating oxidative stress-mediated hepatic dysfunction. The objective of this study was to investigate the mechanisms of the multifunctional effects of cyanidin on regulating antioxidant enzymes and oxidative stress-induced hepatotoxicity. The data indicated that cyanidin-mediated antioxidant enzyme expression involved the extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) pathways and Nrf2 activation. Furthermore, the synergistic effect of cyanidin and the PPAR agonist, troglitazone, on Nrf2-PPAR activation, was also observed. Besides, treatment of cyanidin and troglitazone abolished H₂O₂-induced downregulation of genes involved in lipid metabolism. In addition, H₂O₂-mediated cytotoxicity, which was caused by inducing ROS formation and apoptotic cell death, was also ameliorated upon cyanidin and troglitazone stimulation. In conclusion, mitogen-activated protein kinases (MAPKs) and the transcription factor Nrf2 played regulatory roles in cyanidin-mediated antioxidant enzyme activation. Furthermore, the combination of cyanidin and troglitazone activated PPARγ-Nrf2 and improved H₂O₂-mediated perturbation of genes involved in lipid metabolism. These data suggested that cyanidin and PPAR agonists might have synergistic benefits against metabolic dysfunction-related oxidative damage.
Collapse
Affiliation(s)
- Ping-Hsiao Shih
- Department of Food Science and Biotechnology, National Chung Hsing University, 250 Kuokuang Road, Taichung 40227, Taiwan
| | | | | | | |
Collapse
|
120
|
Polvani S, Tarocchi M, Galli A. PPARγ and Oxidative Stress: Con(β) Catenating NRF2 and FOXO. PPAR Res 2012; 2012:641087. [PMID: 22481913 PMCID: PMC3317010 DOI: 10.1155/2012/641087] [Citation(s) in RCA: 185] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 11/05/2011] [Accepted: 11/17/2011] [Indexed: 12/22/2022] Open
Abstract
Peroxisome-proliferator activator receptor γ (PPARγ) is a nuclear receptor of central importance in energy homeostasis and inflammation. Recent experimental pieces of evidence demonstrate that PPARγ is implicated in the oxidative stress response, an imbalance between antithetic prooxidation and antioxidation forces that may lead the cell to apoptotic or necrotic death. In this delicate and intricate game of equilibrium, PPARγ stands out as a central player devoted to the quenching and containment of the damage and to foster cell survival. However, PPARγ does not act alone: indeed the nuclear receptor is at the point of interconnection of various pathways, such as the nuclear factor erythroid 2-related factor 2 (NRF2), Wnt/β-catenin, and forkhead box proteins O (FOXO) pathways. Here we reviewed the role of PPARγ in response to oxidative stress and its interaction with other signaling pathways implicated in this process, an interaction that emerged as a potential new therapeutic target for several oxidative-related diseases.
Collapse
Affiliation(s)
- Simone Polvani
- Gastroenterology Unit, Department of Clinical Pathophysiology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy
| | - Mirko Tarocchi
- Gastroenterology Unit, Department of Clinical Pathophysiology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy
| | - Andrea Galli
- Gastroenterology Unit, Department of Clinical Pathophysiology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy
| |
Collapse
|
121
|
Tomobe K, Shinozuka T, Kuroiwa M, Nomura Y. Age-related changes of Nrf2 and phosphorylated GSK-3β in a mouse model of accelerated aging (SAMP8). Arch Gerontol Geriatr 2012; 54:e1-7. [DOI: 10.1016/j.archger.2011.06.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 06/01/2011] [Accepted: 06/02/2011] [Indexed: 01/28/2023]
|
122
|
Magri CJ, Gatt N, Xuereb RG, Fava S. Peroxisome proliferator-activated receptor-γ and the endothelium: implications in cardiovascular disease. Expert Rev Cardiovasc Ther 2012; 9:1279-94. [PMID: 21985541 DOI: 10.1586/erc.11.140] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Peroxisome proliferator-activated receptors-γ (PPARγs) are ligand-activated transcription factors that play a crucial regulatory role in the transcription of a large number of genes involved in lipid metabolism and inflammation. In addition to physiological ligands, synthetic ligands (the thiazoledinediones) have been developed. In spite of the much publicized adverse cardiovascular effects of one such thiazoledinedione (rosiglitazone), PPARγ activation may have beneficial cardiovascular effects. In this article we review the effects of PPARγ activation on the endothelium with special emphasis on the possible implications in cardiovascular disease. We discuss its possible role in inflammation, vasomotor function, thrombosis, angiogenesis, vascular aging and vascular rhythm. We also briefly review the clinical implications of these lines of research.
Collapse
Affiliation(s)
- Caroline Jane Magri
- Department of Cardiac Services, Mater Dei Hospital, Tal-Qroqq, Msida MSD 2090, Malta
| | | | | | | |
Collapse
|
123
|
Marcantoni E, Di Francesco L, Totani L, Piccoli A, Evangelista V, Tacconelli S, Patrignani P. Effects of estrogen on endothelial prostanoid production and cyclooxygenase-2 and heme oxygenase-1 expression. Prostaglandins Other Lipid Mediat 2012; 98:122-8. [PMID: 22330859 DOI: 10.1016/j.prostaglandins.2012.01.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Revised: 01/18/2012] [Accepted: 01/27/2012] [Indexed: 11/25/2022]
Abstract
We studied the effects of 17β-estradiol (E₂) (10, 40 nM) on 2 vasoprotective pathways, i.e. cyclooxygenase-2 (COX-2)-dependent prostanoids and the antioxidant heme oxygenase-1 (HO-1), in human umbilical vein endothelial cells (HUVEC) exposed for 6h to steady laminar shear stress (LSS, 10 dyn/cm²), characteristic of atherosclerotic lesion-protected areas. COX-2 was induced by LSS versus static condition (SC). E₂ did not significantly affect COX-2 expression in HUVEC cultured in SC or exposed to LSS. Prostacyclin (PGI₂) and prostaglandin (PG)E₂ were induced while PGF(2α) was reduced by LSS. E₂ caused no effect or a small reduction of prostanoid biosynthesis. In HUVEC cultured in SC or exposed to LSS, E₂ 10 nM caused a comparable HO-1 induction (35-45%) while E₂ 40 nM was 5-fold more potent in LSS-exposed HUVEC than in SC (290% and 58%, respectively). PGI₂ receptor antagonist RO3244794 did not affect HO-1 induction by E₂. In conclusion, E₂ may restrain oxidant stress in the endothelium through HO-1 induction by a mechanism independent on PGI₂ signaling.
Collapse
Affiliation(s)
- Emanuela Marcantoni
- Department of Medicine and Aging, "G. d'Annunzio" University, School of Medicine, 66100 Chieti, Italy
| | | | | | | | | | | | | |
Collapse
|
124
|
Georgiadi A, Boekschoten MV, Müller M, Kersten S. Detailed transcriptomics analysis of the effect of dietary fatty acids on gene expression in the heart. Physiol Genomics 2012; 44:352-61. [PMID: 22274564 DOI: 10.1152/physiolgenomics.00115.2011] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Fatty acids comprise the primary energy source for the heart and are mainly taken up via hydrolysis of circulating triglyceride-rich lipoproteins. While most of the fatty acids entering the cardiomyocyte are oxidized, a small portion is involved in altering gene transcription to modulate cardiometabolic functions. So far, no in vivo model has been developed enabling study of the transcriptional effects of specific fatty acids in the intact heart. In the present study, mice were given a single oral dose of synthetic triglycerides composed of one single fatty acid. Hearts were collected 6 h thereafter and used for whole genome gene expression profiling. Experiments were conducted in wild-type and peroxisome proliferator-activated receptor (PPAR)α-/- mice to allow exploration of the specific contribution of PPARα. It was found that: 1) C18:3 had the most pronounced effect on cardiac gene expression. 2) The largest similarity in gene regulation was observed between C18:2 and C18:3. Large similarity was also observed between PPARα agonist Wy14643 and C22:6. 3) Many genes were regulated by one particular treatment only. Genes regulated by one particular treatment showed large functional divergence. 4) The majority of genes responding to fatty acid treatment were regulated in a PPARα-dependent manner, emphasizing the importance of PPARα in mediating transcriptional regulation by fatty acids in the heart. 5) Several genes were robustly regulated by all or many of the fatty acids studied, mostly representing well-described targets of PPARs (e.g., Acot1, Angptl4, Ucp3) but also including Zbtb16/PLZF, a transcription factor crucial for natural killer T cell function. 6) Deletion and activation of PPARα had a major effect on expression of numerous genes involved in metabolism and immunity. Our analysis demonstrates the marked impact of dietary fatty acids on gene regulation in the heart via PPARα.
Collapse
Affiliation(s)
- Anastasia Georgiadi
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition, Wageningen University, the Netherlands
| | | | | | | |
Collapse
|
125
|
Burgess A, Vanella L, Bellner L, Schwartzman ML, Abraham NG. Epoxyeicosatrienoic acids and heme oxygenase-1 interaction attenuates diabetes and metabolic syndrome complications. Prostaglandins Other Lipid Mediat 2012; 97:1-16. [PMID: 22100745 PMCID: PMC3261364 DOI: 10.1016/j.prostaglandins.2011.10.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Revised: 10/05/2011] [Accepted: 10/17/2011] [Indexed: 12/19/2022]
Abstract
MSCs are considered to be the natural precursors to adipocyte development through the process of adipogenesis. A link has been established between decreased protective effects of EETs or HO-1 and their interaction in metabolic syndrome. Decreases in HO-1 or EET were associated with an increase in adipocyte stem cell differentiation and increased levels of inflammatory cytokines. EET agonist (AKR-I-27-28) inhibited MSC-derived adipocytes and decreased the levels of inflammatory cytokines. We further describe the role of CYP-epoxygenase expression, HO expression, and circulating cytokine levels in an obese mouse, ob/ob(-/-) mouse model. Ex vivo measurements of EET expression within MSCs derived from ob/ob(-/-) showed decreased levels of EETs that were increased by HO induction. This review demonstrates that suppression of HO and EET systems exist in MSCs prior to the development of adipocyte dysfunction. Further, adipocyte dysfunction can be ameliorated by induction of HO-1 and CYP-epoxygenase, i.e. EET.
Collapse
Affiliation(s)
- Angela Burgess
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, OH 43614
| | - Luca Vanella
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, OH 43614
| | - Lars Bellner
- Department of Pharmacology, New York Medical College, Valhalla, NY, 10595
| | | | - Nader G. Abraham
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, OH 43614
| |
Collapse
|
126
|
Sampath S, McLean LA, Buono C, Moulin P, Wolf A, Chibout SD, Pognan F, Busch S, Shangari N, Cruz E, Gurnani M, Patel P, Reising A. The use of rat lens explant cultures to study the mechanism of drug-induced cataractogenesis. Toxicol Sci 2011; 126:128-39. [PMID: 22193206 DOI: 10.1093/toxsci/kfr344] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Lens explant cultures were used to assess the mechanism of drug-induced cataractogenic potential of NVS001, a peroxisome proliferator-activated receptor delta (PPARδ) agonist, which resulted in cataract in all treated animals during a 13-week rat study. Ciglitazone, a PPARγ agonist and cataractogenic compound, was used as a positive control to validate this model. Rat lenses were extracted and cultured in medium supplemented with antibiotics for 24-h preincubation pretreatment. Lenses showing no signs of damage at the end of the preincubation pretreatment period were randomized into five experimental groups, (1) untreated control, (2) 0.1% dimethyl sulphoxide control, (3) 10μM NVS001, (4) 10μM ciglitazone, and (5) 10μM acetaminophen (negative control). Lenses were treated every 24 h after preincubation pretreatment for up to 48 h. Samples for viability, histology, and gene expression profiling were collected at 4, 24, and 48 h. There was a time-dependent increase in opacity, which correlated to a decrease in viability measured by adenosine triphosphate levels in NVS001 and ciglitazone-treated lenses compared with controls. NVS001 and ciglitazone had comparable cataractogenic effects after 48 h with histology showing rupture of the lens capsule, lens fiber degeneration, cortical lens vacuolation, and lens epithelial degeneration. Furthermore, no changes were seen when lenses were treated with acetaminophen. Gene expression analysis supported oxidative and osmotic stress, along with decreases in membrane and epithelial cell integrity as key factors in NVS001-induced cataracts. This study suggests that in vitro lens cultures can be used to assess cataractogenic potential of PPAR agonists and to study/understand the underlying molecular mechanism of cataractogenesis in rat.
Collapse
Affiliation(s)
- Shruthi Sampath
- Investigative Toxicology, Novartis Institutes of Biomedical Research, East Hanover, New Jersey 07936, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
127
|
Cheng CF, Lian WS, Chen SH, Lai PF, Li HF, Lan YF, Cheng WTK, Lin H. Protective effects of adiponectin against renal ischemia-reperfusion injury via prostacyclin-PPARα-heme oxygenase-1 signaling pathway. J Cell Physiol 2011; 227:239-49. [PMID: 21412771 DOI: 10.1002/jcp.22726] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Adiponectin (APN), a circulating adipose-derived hormone that regulates inflammation and energy metabolism, has beneficial effects on the cardiovascular disorders. Serum APN levels are lower in patients with coronary artery disease and higher in patients with chronic kidney disease. However, the precise role of APN in acute reno-vascular disease is not clear. Results of the present study show that serum APN concentration decreased after renal ischemia reperfusion (I/R) injury in mice. In addition, I/R-induced renal dysfunction (elevated serum creatinine and urea levels), inflammation (number of infiltrating neutrophils, myeloperoxidase activity), and apoptotic responses (apoptotic cell number and caspase-3 activation) were attenuated in APN-treated compared to control mice. Molecular and biochemical analysis revealed that APN up-regulates heme oxygenase-1 (HO-1) via peroxisome-proliferator-activated-receptor-α (PPARα) dependent pathway which is mediated through the enhancement of COX-2 and 6-keto PGF1α expression. Chromatin immune-precipitation assay demonstrated that APN increases the binding activity of PPARα to PPRE region of HO-1 promoter. Furthermore, APN induced HO-1 expression was only found in wild-type but not in PPARα gene deleted mice. This provides in vivo evidence that APN mediated HO-1 expression depends on PPARα regulation. In conclusion, our results provide a novel APN mediated prostacyclin-PPARα-HO-1 signaling pathway in protecting renal I/R injury.
Collapse
Affiliation(s)
- Ching-Feng Cheng
- Department of Medical Research, Tzu Chi General Hospital, Hualien, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
128
|
Kadl A, Sharma PR, Chen W, Agrawal R, Meher AK, Rudraiah S, Grubbs N, Sharma R, Leitinger N. Oxidized phospholipid-induced inflammation is mediated by Toll-like receptor 2. Free Radic Biol Med 2011; 51:1903-9. [PMID: 21925592 PMCID: PMC3197756 DOI: 10.1016/j.freeradbiomed.2011.08.026] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 08/22/2011] [Accepted: 08/23/2011] [Indexed: 12/18/2022]
Abstract
Oxidative tissue damage is a hallmark of many chronic inflammatory diseases. However, the precise mechanisms linking oxidative changes to inflammatory reactions remain unclear. Herein we show that Toll-like receptor 2 (TLR2) translates oxidative tissue damage into inflammatory responses by mediating the effects of oxidized phospholipids. Intraperitoneal injection of oxidized 1-palmitoyl-2-arachidonyl-sn-3-glycerophosphorylcholine (OxPAPC) resulted in upregulation of inflammatory genes in wild-type, but not in TLR2(-/-) mice. In vitro, OxPAPC induced TLR2 (but not TLR4)-dependent inflammatory gene expression and JNK and p38 signaling in macrophages. Induction of TLR2-dependent gene expression required reducible functional groups on sn-2 acyl chains of oxidized phospholipids, as well as serum cofactors. Finally, TLR2(-/-) mice were protected against carbon tetrachloride-induced oxidative tissue damage and inflammation, which was accompanied by accumulation of oxidized phospholipids in livers. Together, our findings demonstrate that TLR2 mediates cellular responses to oxidative tissue damage and they provide new insights into how oxidative stress is linked to acute and chronic inflammation.
Collapse
Affiliation(s)
- Alexandra Kadl
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Poonam R. Sharma
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Wenshu Chen
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Rachana Agrawal
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Akshaya K. Meher
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Swetha Rudraiah
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Nathaniel Grubbs
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Rahul Sharma
- Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Norbert Leitinger
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
129
|
McCarthy FP, Drewlo S, English FA, Kingdom J, Johns EJ, Kenny LC, Walsh SK. Evidence Implicating Peroxisome Proliferator-Activated Receptor-γ in the Pathogenesis of Preeclampsia. Hypertension 2011; 58:882-7. [DOI: 10.1161/hypertensionaha.111.179440] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Preeclampsia, a major cause of maternal and perinatal mortality and morbidity, is thought to be attributed, in part, to impaired trophoblast invasion. Peroxisome proliferator-activated receptors are ligand-activated transcription factors expressed in trophoblasts, which regulate the expression of a number of genes involved in cell differentiation and proliferation. We investigated the effect of the administration of a peroxisome proliferator-activated receptor-γ antagonist during uncomplicated pregnancy in rats. Using an intraperitoneal miniosmotic pump, healthy pregnant rats were administered either vehicle or the peroxisome proliferator-activated receptor-γ–specific antagonist, T0070907 (1 mg/kg per day from gestational days 11–15). Rats treated with T0070907 developed key features of preeclampsia, including elevated mean arterial blood pressure, proteinuria, endothelial dysfunction, reduced pup weight, and increased platelet aggregation. T0070907-treated rats had reduced plasma vascular endothelial growth factor and increased plasma soluble fms-like tyrosine kinase 1. Furthermore, increases in total placental soluble fms-like tyrosine kinase 1 mRNA and fms-like tyrosine kinase 1 protein were also demonstrated, suggesting the placenta as the main contributor to the increased circulating levels of soluble fms-like tyrosine kinase 1. The labyrinthine trophoblast in the placentas of T0070907-treated rats were less differentiated, had increased cellular proliferation, and were strongly immunopositive for CD-31 staining, indicating adaptive angiogenesis. The present study suggests that peroxisome proliferator-activated receptor-γ may play a pivotal role in the progression of a healthy pregnancy and may critically regulate the risk of preeclampsia. These findings have important implications regarding the underlying etiology of preeclampsia and potential therapeutic targets.
Collapse
Affiliation(s)
- Fergus P. McCarthy
- From the Anu Research Centre, Department of Obstetrics and Gynaecology (F.P.M., F.A.E., L.C.K., S.K.W.), and Department of Physiology (E.J.J.), University College Cork, Cork, Ireland; Samuel Lunenfeld Research Institute (S.D., J.K.), Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Sascha Drewlo
- From the Anu Research Centre, Department of Obstetrics and Gynaecology (F.P.M., F.A.E., L.C.K., S.K.W.), and Department of Physiology (E.J.J.), University College Cork, Cork, Ireland; Samuel Lunenfeld Research Institute (S.D., J.K.), Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Fred A. English
- From the Anu Research Centre, Department of Obstetrics and Gynaecology (F.P.M., F.A.E., L.C.K., S.K.W.), and Department of Physiology (E.J.J.), University College Cork, Cork, Ireland; Samuel Lunenfeld Research Institute (S.D., J.K.), Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - John Kingdom
- From the Anu Research Centre, Department of Obstetrics and Gynaecology (F.P.M., F.A.E., L.C.K., S.K.W.), and Department of Physiology (E.J.J.), University College Cork, Cork, Ireland; Samuel Lunenfeld Research Institute (S.D., J.K.), Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Edward J. Johns
- From the Anu Research Centre, Department of Obstetrics and Gynaecology (F.P.M., F.A.E., L.C.K., S.K.W.), and Department of Physiology (E.J.J.), University College Cork, Cork, Ireland; Samuel Lunenfeld Research Institute (S.D., J.K.), Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Louise C. Kenny
- From the Anu Research Centre, Department of Obstetrics and Gynaecology (F.P.M., F.A.E., L.C.K., S.K.W.), and Department of Physiology (E.J.J.), University College Cork, Cork, Ireland; Samuel Lunenfeld Research Institute (S.D., J.K.), Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Sarah K. Walsh
- From the Anu Research Centre, Department of Obstetrics and Gynaecology (F.P.M., F.A.E., L.C.K., S.K.W.), and Department of Physiology (E.J.J.), University College Cork, Cork, Ireland; Samuel Lunenfeld Research Institute (S.D., J.K.), Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
130
|
Fei J, Cook C, Gillespie M, Yu B, Fullen K, Santanam N. Atherogenic ω-6 Lipids Modulate PPAR- EGR-1 Crosstalk in Vascular Cells. PPAR Res 2011; 2011:753917. [PMID: 22135674 PMCID: PMC3205716 DOI: 10.1155/2011/753917] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2011] [Revised: 06/17/2011] [Accepted: 07/11/2011] [Indexed: 02/03/2023] Open
Abstract
Atherogenic ω-6 lipids are physiological ligands of peroxisome proliferator-activated receptors (PPARs) and elicit pro- and antiatherogenic responses in vascular cells. The objective of this study was to investigate if ω-6 lipids modulated the early growth response-1 (Egr-1)/PPAR crosstalk thereby altering vascular function. Rat aortic smooth muscle cells (RASMCs) were exposed to ω-6 lipids, linoleic acid (LA), or its oxidized form, 13-HPODE (OxLA) in the presence or absence of a PPARα antagonist (MK886) or PPARγ antagonist (GW9662) or PPAR-specific siRNA. Our results demonstrate that ω-6 lipids, induced Egr-1 and monocyte chemotactic protein-1 (MCP-1) mRNA and protein levels at the acute phase (1-4 hrs) when PPARα was downregulated and at subacute phase (4-12 hrs) by modulating PPARγ, thus resulting in altered monocyte adhesion to RASMCs. We provide novel insights into the mechanism of action of ω-6 lipids on Egr-1/PPAR interactions in vascular cells and their potential in altering vascular function.
Collapse
Affiliation(s)
- Jia Fei
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Carla Cook
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Miriah Gillespie
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Bangning Yu
- Department of Pharmacology and Experimental Therapeutics, LSU Health Sciences Center, New Orleans, LA, USA
| | - Khyra Fullen
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| | - Nalini Santanam
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA
| |
Collapse
|
131
|
von Knethen A, Neb H, Morbitzer V, Schmidt MV, Kuhn AM, Kuchler L, Brüne B. PPARγ stabilizes HO-1 mRNA in monocytes/macrophages which affects IFN-β expression. Free Radic Biol Med 2011; 51:396-405. [PMID: 21571064 DOI: 10.1016/j.freeradbiomed.2011.04.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Revised: 03/29/2011] [Accepted: 04/18/2011] [Indexed: 01/02/2023]
Abstract
NADPH oxidase activation in either RAW264.7 cells or peritoneal macrophages (PM) derived from PPARγ wild-type mice increased reactive oxygen species (ROS) formation, caused PPARγ activation, heme oxygenase-1 (HO-1) induction, and concomitant IFN-β expression. In macrophages transduced with a dominant negative (d/n) mutant of PPARγ (RAW264.7 AF2) as well as PPARγ negative PM derived from Mac-PPARγ-KO mice, NADPH oxidase-dependent IFN-β expression was attenuated. As the underlying mechanism, we noted decreased HO-1 mRNA stability in RAW264.7 AF2 cells as well as PPARγ negative PM, compared to either parent RAW264.7 cells or wild-type PM. Assuming mRNA stabilization of HO-1 by PPARγ we transfected macrophages with a HO-1 3'-UTR reporter construct. The PPARγ agonist rosiglitazone significantly up-regulated luciferase expression in RAW264.7 cells, while it remained unaltered in RAW264.7 AF2 macrophages. Deletion of each of two AU-rich elements in the 3'-UTR HO-1 decreased luciferase activity in RAW264.7 cells. Using LPS as a NADPH oxidase activator, PM from Mac-PPARγ-KO mice showed a decreased HO-1 mRNA half-life in vitro and in vivo compared to PPARγ wild-type mice. These data identified a so far unappreciated role of PPARγ in stabilizing HO-1 mRNA, thus, contributing to the expression of the HO-1 target gene IFN-β.
Collapse
Affiliation(s)
- Andreas von Knethen
- Institute of Biochemistry I-Pathobiochemistry, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Theodor-Stern-Kai 7, Germany.
| | | | | | | | | | | | | |
Collapse
|
132
|
Lissauer M, Feild C, Fasanmi F, Johnson SB. Rosiglitazone may assist with glycemic control in the ICU. J Intensive Care Med 2011; 25:117-20. [PMID: 19955116 DOI: 10.1177/0885066609355264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Hyperglycemia is a frequent sequela of critical illness. Rosiglitazone is an oral hypoglycemic agent of the thiazolinedione class. Thiazolinediones are known to activate peroxisome proliferator-activated receptor gamma (PPAR-gamma) that decreases inflammation in humans and decreases shock induced by zymosan in mice. HYPOTHESIS Rosiglitazone can assist with hyperglycemic control in the intensive care unit (ICU). METHODS A hospital billing query identified patients prescribed rosiglitazone while in a major university ICU. Patients who received rosiglitazone as an outpatient prior to hospitalization were excluded. Glycemic control was determined by average daily blood glucose, 24-hour insulin dose, and number of patients requiring an insulin drip. Glycemic control was evaluated on days 0, 3, and 7. Student t test was used to compare means. Fisher exact testing was used to compare insulin regimen before and after starting rosiglitazone. RESULTS 34 patients were identified. The average Acute Physiology and Chronic Health Evaluation (APACHE) II score was 17.2 +/- 4.4. Sixty-five percent were male, 62% were preexisting diabetics. The mean daily blood glucose was 159 +/- 30 mg/dL on day 0, 146 +/- 37 mg/dL on day 3, and 140 +/- 33 mg/dL on day 7 (P < .03 vs day 0). The mean 24-hour insulin dose was 80.6 +/- 87.9 U on day 0, 72.2 +/- 73.4 U on day 3, and 46.3 +/- 57.2 U on day 7 (P < .003 vs day 0). There was 1 major hypoglycemic event. CONCLUSION Rosiglitazone may assist glycemic control in the ICU. Despite recent concerns of cardiac safety, further research should be done to evaluate its potential as a short-term therapeutic agent in the ICU, given its anti-inflammatory and antishock profile.
Collapse
Affiliation(s)
- Matthew Lissauer
- Department of Surgical Critical Care, University of Maryland Medical Center, R Adams Cowley Shock Trauma Center, Baltimore, Maryland, USA.
| | | | | | | |
Collapse
|
133
|
McCarthy FP, Drewlo S, Kingdom J, Johns EJ, Walsh SK, Kenny LC. Peroxisome proliferator-activated receptor-γ as a potential therapeutic target in the treatment of preeclampsia. Hypertension 2011; 58:280-6. [PMID: 21690483 DOI: 10.1161/hypertensionaha.111.172627] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Preeclampsia is a multisystemic disorder of pregnancy characterized by hypertension, proteinuria, and maternal endothelial dysfunction. It is a major cause of maternal and perinatal morbidity and mortality and is thought to be attributable, in part, to inadequate trophoblast invasion. Peroxisome proliferator-activated receptor-γ (PPAR-γ) is a ligand-activated transcription factor expressed in trophoblasts, and the vasculature of which activation has been shown to improve endothelium-dependent vasodilatation in hypertensive conditions. We investigated the effects of the administration of a PPAR-γ agonist using the reduced uterine perfusion pressure (RUPP) rat model of preeclampsia. The selective PPAR-γ agonist, rosiglitazone, was administered to pregnant rats that had undergone RUPP surgery. To investigate whether any observed beneficial effects of PPAR-γ activation were mediated by the antioxidant enzyme, heme oxygenase 1, rosiglitazone was administered in combination with the heme oxygenase 1 inhibitor tin-protoporphyrin IX. RUPP rats were characterized by hypertension, endothelial dysfunction, and elevated microalbumin:creatinine ratios. Rosiglitazone administration ameliorated hypertension, improved vascular function, and reduced the elevated microalbumin:creatinine ratio in RUPP rats. With the exception of microalbumin:creatinine ratio, these beneficial effects were abrogated in the presence of the heme oxygenase 1 inhibitor. Administration of a PPAR-γ agonist prevented the development of several of the pathophysiological characteristics associated with the RUPP model of preeclampsia, via a heme oxygenase 1-dependent pathway. The findings from this study provide further insight into the underlying etiology of preeclampsia and a potential therapeutic target for the treatment of preeclampsia.
Collapse
Affiliation(s)
- Fergus P McCarthy
- Anu Research Centre, University College Cork, Cork University Maternity Hospital, Wilton, Cork, Ireland.
| | | | | | | | | | | |
Collapse
|
134
|
Serghides L. The Case for the Use of PPARγ Agonists as an Adjunctive Therapy for Cerebral Malaria. PPAR Res 2011; 2012:513865. [PMID: 21772838 PMCID: PMC3135089 DOI: 10.1155/2012/513865] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Accepted: 02/28/2011] [Indexed: 12/24/2022] Open
Abstract
Cerebral malaria is a severe complication of Plasmodium falciparum infection associated with high mortality even when highly effective antiparasitic therapy is used. Adjunctive therapies that modify the pathophysiological processes caused by malaria are a possible way to improve outcome. This review focuses on the utility of PPARγ agonists as an adjunctive therapy for the treatment of cerebral malaria. The current knowledge of PPARγ agonist use in malaria is summarized. Findings from experimental CNS injury and disease models that demonstrate the potential for PPARγ agonists as an adjunctive therapy for cerebral malaria are also discussed.
Collapse
Affiliation(s)
- Lena Serghides
- Sandra A. Rotman Laboratories, McLaughlin-Rotman Centre for Global Health, Toronto General Hospital, University Health Network, 101 College Street, Suite 10-359, Toronto, ON, Canada M5G 1L7
| |
Collapse
|
135
|
Park SY, Bae JU, Hong KW, Kim CD. HO-1 Induced by Cilostazol Protects Against TNF-α-associated Cytotoxicity via a PPAR-γ-dependent Pathway in Human Endothelial Cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2011; 15:83-8. [PMID: 21660147 DOI: 10.4196/kjpp.2011.15.2.83] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2011] [Revised: 03/10/2011] [Accepted: 03/14/2011] [Indexed: 12/27/2022]
Abstract
A large body of evidence has indicated that induction of endogenous antioxidative proteins seems to be a reasonable strategy for delaying the progression of cell injury. In our previous study, cilostazol was found to increase the expression of the antioxidant enzyme heme oxygenase-1 (HO-1) in synovial cells. Thus, the present study was undertaken to examine whether cilostazol is able to counteract tumor necrosis factor-α (TNF-α)-induced cell death in endothelial cells via the induction of HO-1 expression. We exposed human umbilical vein endothelial cells (HUVECs) to TNF-α (50 ng/ml), with or without cilostazol (10 µM). Pretreatment with cilostazol markedly reduced TNF-α-induced viability loss in the HUVECs, which was reversed by zinc protoporphyrine IX (ZnPP), an inhibitor of HO-1. Moreover, cilostazol increased HO-1 protein and mRNA expression. Cilostazol-induced HO-1 induction was markedly attenuated not only by ZnPP but also by copper-protoporphyrin IX (CuPP). In an assay measuring peroxisome proliferator-activated receptor-γ (PPAR-γ) transcription activity, cilostazol directly increased PPAR-γ transcriptional activity which was completely abolished by HO-1 inhibitor. Furthermore, increased PPAR-γ activity by cilostazol and rosiglitazone was completely abolished in cells transfected with HO-1 siRNA. Taken together, these results indicate that cilostazol up-regulates HO-1 and protects cells against TNF-α-induced endothelial cytotoxicity via a PPAR-γ-dependent pathway.
Collapse
Affiliation(s)
- So Youn Park
- Department of Pharmacology, School of Medicine, and MRC for Ischemic Tissue Regeneration and Medical Research Institute, Pusan National University, Yangsan 626-770, Korea
| | | | | | | |
Collapse
|
136
|
Roos TU, Heiss EH, Schwaiberger AV, Schachner D, Sroka IM, Oberan T, Vollmar AM, Dirsch VM. Caffeic acid phenethyl ester inhibits PDGF-induced proliferation of vascular smooth muscle cells via activation of p38 MAPK, HIF-1α, and heme oxygenase-1. JOURNAL OF NATURAL PRODUCTS 2011; 74:352-356. [PMID: 21265554 DOI: 10.1021/np100724f] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Hyperproliferation of vascular smooth muscle cells (VSMCs) is critically involved in the onset of atherosclerosis and restenosis. Although caffeic acid phenethyl ester (CAPE, 1), one of the main constituents of honeybee propolis, has been shown to exert a beneficial effect in models of vascular injury in vivo, detailed mechanistic investigations in vascular cells are scarce. This study has examined the antiproliferative activity of 1 in platelet-derived growth factor (PDGF)-stimulated primary rat aortic VSMCs and aimed to shed light on underlying molecular mechanisms. Compound 1 inhibited the proliferation of VSMCs upon exposure to PDGF in a dose-dependent manner by interfering with cell cycle progression from the G0/1- to the S-phase. Enhanced phosphorylation of p38 mitogen-activated protein kinase (MAPK) as well as stabilization of hypoxia-inducible factor (HIF)-1α and subsequent induction of heme oxygenase-1 (HO-1) could be identified as molecular events contributing to the observed growth arrest in PDGF-activated VSMCs upon exposure to 1.
Collapse
Affiliation(s)
- Thomas U Roos
- Department of Pharmacy, Center of Drug Research, University of Munich, 81377 Munich, Germany
| | | | | | | | | | | | | | | |
Collapse
|
137
|
Kim HJ, Ham SA, Paek KS, Hwang JS, Jung SY, Kim MY, Jin H, Kang ES, Woo IS, Kim HJ, Lee JH, Chang KC, Han CW, Seo HG. Transcriptional up-regulation of antioxidant genes by PPARδ inhibits angiotensin II-induced premature senescence in vascular smooth muscle cells. Biochem Biophys Res Commun 2011; 406:564-9. [PMID: 21352808 DOI: 10.1016/j.bbrc.2011.02.091] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Accepted: 02/18/2011] [Indexed: 12/14/2022]
Abstract
This study evaluated peroxisome proliferator-activated receptor (PPAR) δ as a potential target for therapeutic intervention in Ang II-induced senescence in human vascular smooth muscle cells (hVSMCs). Activation of PPARδ by GW501516, a specific agonist of PPARδ, significantly inhibited the Ang II-induced premature senescence of hVSMCs. Agonist-activated PPARδ suppressed the generation of Ang II-triggered reactive oxygen species (ROS) with a concomitant reduction in DNA damage. Notably, GW501516 up-regulated the expression of antioxidant genes, such as glutathione peroxidase 1, thioredoxin 1, manganese superoxide dismutase and heme oxygenase 1. siRNA-mediated down-regulation of these antioxidant genes almost completely abolished the effects of GW501516 on ROS production and premature senescence in hVSMCs treated with Ang II. Taken together, the enhanced transcription of antioxidant genes is responsible for the PPARδ-mediated inhibition of premature senescence through sequestration of ROS in hVSMCs treated with Ang II.
Collapse
Affiliation(s)
- Hyo Jung Kim
- Department of Pharmacology, Gyeongsang Institute of Health Science, Gyeongsang National University School of Medicine, Jinju, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
138
|
Doberer D, Haschemi A, Andreas M, Zapf TC, Clive B, Jeitler M, Heinzl H, Wagner O, Wolzt M, Bilban M. Haem arginate infusion stimulates haem oxygenase-1 expression in healthy subjects. Br J Pharmacol 2011; 161:1751-62. [PMID: 20718734 DOI: 10.1111/j.1476-5381.2010.00990.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Haem oxygenase 1 (HO-1) is an inducible protein that plays a major protective role in conditions such as ischaemia-reperfusion injury and inflammation. In this study, we have investigated the role of haem arginate (HA) in human male subjects in the modulation of HO-1 expression and its correlation with the GT length polymorphism (GT(n)) in the promoter of the HO-1 gene. EXPERIMENTAL APPROACH In a dose-escalation, randomized, placebo-controlled trial, seven healthy male subjects with a homozygous short (S/S) and eight with a long (L/L) GT(n) genotype received intravenous HA. HO-1 protein expression and mRNA levels in peripheral blood monocytes, bilirubin, haptoglobin, haemopexin and haem levels were analysed over a 48 h observation period. KEY RESULTS We found that the baseline mRNA levels of HO-1 were higher in L/L subjects, while protein levels were higher in S/S subjects. HA induced a dose-dependent increase in the baseline corrected area under the curve values of HO-1 mRNA and protein over 48 h. The response of HO-1 mRNA was more pronounced in L/L subjects but the protein level was similar across the groups. CONCLUSIONS AND IMPLICATION HA is an effective inducer of HO-1 in humans irrespective of the GT(n) genotype. The potential therapeutic application of HA needs to be evaluated in clinical trials.
Collapse
Affiliation(s)
- D Doberer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
139
|
Haem oxygenase-1 and cardiovascular disease: mechanisms and therapeutic potential. Clin Sci (Lond) 2011; 120:493-504. [DOI: 10.1042/cs20100508] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Cardiovascular disease remains the leading cause of death worldwide. Despite progress in management, there remain a significant number of patients who are not eligible for current treatment options. Traditionally, HO-1 (haem oxygenase-1), one of two isoenzymes that initiate haem catabolism, was thought to only play a metabolic role. However, HO-1 is now recognized to have additional protective activities in states of heightened noxious stimuli or stress such as acute coronary syndromes. The present review article provides an overview of the mode of action of HO-1 in vascular protection, with particular emphasis on its atheroprotective, anti-inflammatory and antioxidative properties, as well as its role in vascular repair. Furthermore, we present evidence for the protective effects of HO-1 in CVD (cardiovascular disease) in both animal and human studies. Given its potential in vascular protection and repair, strategies aimed at inducing HO-1 emerge as a novel and alternative therapeutic target in the management of CVD.
Collapse
|
140
|
|
141
|
Rayman MP, Wijnen H, Vader H, Kooistra L, Pop V. Maternal selenium status during early gestation and risk for preterm birth. CMAJ 2011; 183:549-55. [PMID: 21324870 DOI: 10.1503/cmaj.101095] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Preterm birth occurs in 5%-13% of pregnancies. It is a leading cause of perinatal mortality and morbidity and has adverse long-term consequences for the health of the child. Because of the role selenium plays in attenuating inflammation, and because low concentrations of selenium have been found in women with preeclampsia, we hypothesized that low maternal selenium status during early gestation would increase the risk of preterm birth. METHODS White Dutch women with a singleton pregnancy (n = 1197) were followed prospectively from 12 weeks' gestation. Women with thyroid disease or type 1 diabetes were excluded. At delivery, 1129 women had complete birth-outcome data. Serum concentrations of selenium were measured during the 12th week of pregnancy. Deliveries were classified as preterm or term, and preterm births were subcategorized as iatrogenic, spontaneous or the result of premature rupture of the membranes. RESULTS Of the 60 women (5.3%) who had a preterm birth, 21 had premature rupture of the membranes and 13 had preeclampsia. The serum selenium concentration at 12 weeks' gestation was significantly lower among women who had a preterm birth than among those who delivered at term (mean 0.96 [standard deviation (SD) 0.14] μmol/L v. 1.02 [SD 0.13] μmol/L; t = 2.9, p = 0.001). Women were grouped by quartile of serum selenium concentration at 12 weeks' gestation. The number of women who had a preterm birth significantly differed by quartile (χ² = 8.01, 3 degrees of freedom], p < 0.05). Women in the lowest quartile of serum selenium had twice the risk of preterm birth as women in the upper three quartiles, even after adjustment for the occurrence of preeclampsia (adjusted odds ratio 2.18, 95% confidence interval 1.25-3.77). INTERPRETATION Having low serum selenium at the end of the first trimester was related to preterm birth and was independent of the mother having preeclampsia. Low maternal selenium status during early gestation may increase the risk of preterm premature rupture of the membranes, which is a major cause of preterm birth.
Collapse
Affiliation(s)
- Margaret P Rayman
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK.
| | | | | | | | | |
Collapse
|
142
|
Wu QQ, Wang Y, Senitko M, Meyer C, Wigley WC, Ferguson DA, Grossman E, Chen J, Zhou XJ, Hartono J, Winterberg P, Chen B, Agarwal A, Lu CY. Bardoxolone methyl (BARD) ameliorates ischemic AKI and increases expression of protective genes Nrf2, PPARγ, and HO-1. Am J Physiol Renal Physiol 2011; 300:F1180-92. [PMID: 21289052 DOI: 10.1152/ajprenal.00353.2010] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Ischemic acute kidney injury (AKI) triggers expression of adaptive (protective) and maladaptive genes. Agents that increase expression of protective genes should provide a therapeutic benefit. We now report that bardoxolone methyl (BARD) ameliorates ischemic murine AKI as assessed by both renal function and pathology. BARD may exert its beneficial effect by increasing expression of genes previously shown to protect against ischemic AKI, NF-E2-related factor 2 (Nrf2), peroxisome proliferator-activated receptor-γ (PPARγ), and heme oxygenase 1 (HO-1). Although we found that BARD alone or ischemia-reperfusion alone increased expression of these genes, the greatest increase occurred after the combination of both ischemia-reperfusion and BARD. BARD had a different mode of action than other agents that regulate PPARγ and Nrf2. Thus we report that BARD regulates PPARγ, not by acting as a ligand but by increasing the amount of PPARγ mRNA and protein. This should increase ligand-independent effects of PPARγ. Similarly, BARD increased Nrf2 mRNA; this increased Nrf2 protein by mechanisms in addition to the prolongation of Nrf2 protein half-life previously reported. Finally, we localized expression of these protective genes after ischemia and BARD treatment. Using double-immunofluorescence staining for CD31 and Nrf2 or PPARγ, we found increased Nrf2 and PPARγ on glomerular endothelia in the cortex; Nrf2 was also present on cortical peritubular capillaries. In contrast, HO-1 was localized to different cells, i.e., tubules and interstitial leukocytes. Although Nrf2-dependent increases in HO-1 have been described, our data suggest that BARD's effects on tubular and leukocyte HO-1 during ischemic AKI may be Nrf2 independent. We also found that BARD ameliorated cisplatin nephrotoxicity.
Collapse
Affiliation(s)
- Qing Qing Wu
- Department of Internal Medicine, Nephrology Division, UT Southwestern Medical Center, Dallas, TX 75390-8856, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Kim YM, Pae HO, Park JE, Lee YC, Woo JM, Kim NH, Choi YK, Lee BS, Kim SR, Chung HT. Heme oxygenase in the regulation of vascular biology: from molecular mechanisms to therapeutic opportunities. Antioxid Redox Signal 2011; 14:137-67. [PMID: 20624029 PMCID: PMC2988629 DOI: 10.1089/ars.2010.3153] [Citation(s) in RCA: 181] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Heme oxygenases (HOs) are the rate-limiting enzymes in the catabolism of heme into biliverdin, free iron, and carbon monoxide. Two genetically distinct isoforms of HO have been characterized: an inducible form, HO-1, and a constitutively expressed form, HO-2. HO-1 is a kind of stress protein, and thus regarded as a sensitive and reliable indicator of cellular oxidative stress. The HO system acts as potent antioxidants, protects endothelial cells from apoptosis, is involved in regulating vascular tone, attenuates inflammatory response in the vessel wall, and participates in angiogenesis and vasculogenesis. Endothelial integrity and activity are thought to occupy the central position in the pathogenesis of cardiovascular diseases. Cardiovascular disease risk conditions converge in the contribution to oxidative stress. The oxidative stress leads to endothelial and vascular smooth muscle cell dysfunction with increases in vessel tone, cell growth, and gene expression that create a pro-thrombotic/pro-inflammatory environment. Subsequent formation, progression, and obstruction of atherosclerotic plaque may result in myocardial infarction, stroke, and cardiovascular death. This background provides the rationale for exploring the potential therapeutic role for HO system in the amelioration of vascular inflammation and prevention of adverse cardiovascular outcomes.
Collapse
Affiliation(s)
- Young-Myeong Kim
- Vascular System Research Center and Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Kangwon-do, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
144
|
Kim YM, Pae HO, Park JE, Lee YC, Woo JM, Kim NH, Choi YK, Lee BS, Kim SR, Chung HT. Heme oxygenase in the regulation of vascular biology: from molecular mechanisms to therapeutic opportunities. Antioxid Redox Signal 2010. [PMID: 20624029 DOI: 10.1089/ars.2010.31532988629] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Heme oxygenases (HOs) are the rate-limiting enzymes in the catabolism of heme into biliverdin, free iron, and carbon monoxide. Two genetically distinct isoforms of HO have been characterized: an inducible form, HO-1, and a constitutively expressed form, HO-2. HO-1 is a kind of stress protein, and thus regarded as a sensitive and reliable indicator of cellular oxidative stress. The HO system acts as potent antioxidants, protects endothelial cells from apoptosis, is involved in regulating vascular tone, attenuates inflammatory response in the vessel wall, and participates in angiogenesis and vasculogenesis. Endothelial integrity and activity are thought to occupy the central position in the pathogenesis of cardiovascular diseases. Cardiovascular disease risk conditions converge in the contribution to oxidative stress. The oxidative stress leads to endothelial and vascular smooth muscle cell dysfunction with increases in vessel tone, cell growth, and gene expression that create a pro-thrombotic/pro-inflammatory environment. Subsequent formation, progression, and obstruction of atherosclerotic plaque may result in myocardial infarction, stroke, and cardiovascular death. This background provides the rationale for exploring the potential therapeutic role for HO system in the amelioration of vascular inflammation and prevention of adverse cardiovascular outcomes.
Collapse
Affiliation(s)
- Young-Myeong Kim
- Vascular System Research Center and Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Kangwon-do, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
145
|
Kim JW, Lim SC, Lee MY, Lee JW, Oh WK, Kim SK, Kang KW. Inhibition of neointimal formation by trans-resveratrol: role of phosphatidyl inositol 3-kinase-dependent Nrf2 activation in heme oxygenase-1 induction. Mol Nutr Food Res 2010; 54:1497-1505. [PMID: 20486211 DOI: 10.1002/mnfr.201000016] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Neointima, defined as abnormal growth of the intimal layer of blood vessels, is believed to be a critical event in the development of vascular occlusive disease. Although resveratrol's inhibitory effects on proliferation and migration of vascular smooth muscle cells has been reported, its activity on neointimal formation is still unclear. Oral administration of trans-resveratrol significantly suppressed intimal hyperplasia in a wire-injured femoral artery mouse model. In cultured vascular smooth muscle cells, trans-resveratrol inhibited platelet-derived growth factor-stimulated DNA synthesis and cell proliferation with down-regulation of cyclin D and pRB. Moreover, platelet-derived growth factor-induced production of reactive oxygen species was inhibited by trans-resveratrol and the compound induced heme oxygenase-1 (HO-1). The anti-proliferative activity of trans-resveratrol was reversed by an HO-1 inhibitor, ZnPPIX. Subcellular fractionation and reporter gene analyses revealed that trans-resveratrol increased the level of nuclear Nrf2 and antioxidant response element reporter activity, and that these were essential for the induction of HO-1. Trans-resveratrol also enhanced the activities of phosphatidyl inositol 3-kinase and extracellular signal regulated kinase, and phosphatidyl inositol 3-kinase was required for Nrf2/antioxidant response element-dependent HO-1 induction. These data have significant implications for the elucidation of the pharmacological mechanism by which trans-resveratrol prevents vascular occlusive diseases.
Collapse
Affiliation(s)
- Jung Woo Kim
- BK21 Project Team, College of Pharmacy, Chosun University, Gwangju, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
146
|
Abstract
Metabolic syndrome (MetS) is a constellation of risk factors including insulin resistance, central obesity, dyslipidemia and hypertension that markedly increase the risk of Type 2 diabetes (T2DM) and cardiovascular disease (CVD). The peroxisome proliferators-activated receptor (PPAR) isotypes, PPARα, PPARδ/ß and PPARγ are ligand-activated nuclear transcription factors, which modulate the expression of an array of genes that play a central role in regulating glucose, lipid and cholesterol metabolism, where imbalance can lead to obesity, T2DM and CVD. They are also drug targets, and currently, PPARα (fibrates) and PPARγ (thiazolodinediones) agonists are in clinical use for treating dyslipidemia and T2DM, respectively. These metabolic characteristics of the PPARs, coupled with their involvement in metabolic diseases, mean extensive efforts are underway worldwide to develop new and efficacious PPAR-based therapies for the treatment of additional maladies associated with the MetS. This article presents an overview of the functional characteristics of three PPAR isotypes, discusses recent advances in our understanding of the diverse biological actions of PPARs, particularly in the vascular system, and summarizes the developmental status of new single, dual, pan (multiple) and partial PPAR agonists for the clinical management of key components of MetS, T2DM and CVD. It also summarizes the clinical outcomes from various clinical trials aimed at evaluating the atheroprotective actions of currently used fibrates and thiazolodinediones.
Collapse
Affiliation(s)
- Salman Azhar
- Geriatric Research, Education & Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA, USA.
| |
Collapse
|
147
|
Pae HO, Son Y, Kim NH, Jeong HJ, Chang KC, Chung HT. Role of heme oxygenase in preserving vascular bioactive NO. Nitric Oxide 2010; 23:251-7. [PMID: 20713168 DOI: 10.1016/j.niox.2010.08.002] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2009] [Revised: 07/22/2010] [Accepted: 08/11/2010] [Indexed: 01/12/2023]
Abstract
Beyond its vasodilator role, vascular nitric oxide (NO), which is synthesized by endothelial NO synthase (eNOS) via its activation, has been shown to play a number of other beneficial roles in the vascular system; it inhibits proliferation of vascular smooth muscle cells, prevents platelet aggregation, and regulates endothelial apoptosis. Such beneficial roles have been shown to be implicated in the regulation of endothelial functions. A loss of NO bioavailability that may result either from decreased eNOS expression and activity or from increased NO degradation is associated with endothelial dysfunction, a key factor in the development of vascular diseases. Heme oxygenase-1 (HO-1), an inducible enzyme, catalyzes the oxidative degradation of heme to free iron, carbon monoxide, and biliverdin, the latter being subsequently converted into bilirubin. In the vascular system, HO-1 and heme degradation products perform important physiological functions, which are ultimately linked to the protection of vascular cells. Studies have shown that HO-1 and heme degradation products exert vasodilatory, antioxidant, anti-inflammatory, antiproliferative and anti-apoptotic effects on vascular cells. Interestingly, these effects of HO-1 and its by-products are similar, at least in part, to those of eNOS-derived NO; this similarity may prompt investigators to study a possible relationship between eNOS-derived NO and HO-1 pathways. Many studies have been reported, and accumulating evidence suggests that HO-1 and heme degradation products can improve vascular function, at least in part, by compensating for the loss of NO bioavailability. This paper will provide the possible pathway explaining how HO-1 and heme degradation products can preserve vascular NO.
Collapse
Affiliation(s)
- Hyun-Ock Pae
- Department of Microbiology and Immunology, Wonkwang University School of Medicine, Iksan 570-749, Republic of Korea
| | | | | | | | | | | |
Collapse
|
148
|
Lin H, Yu CH, Jen CY, Cheng CF, Chou Y, Chang CC, Juan SH. Adiponectin-mediated heme oxygenase-1 induction protects against iron-induced liver injury via a PPARα dependent mechanism. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:1697-709. [PMID: 20709802 DOI: 10.2353/ajpath.2010.090789] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Protective effects of adiponectin (APN; an adipocytokine) were shown against various oxidative challenges; however, its therapeutic implications and the mechanisms underlying hepatic iron overload remain unclear. Herein, we show that the deleterious effects of iron dextran on liver function and iron deposition were significantly reversed by adiponectin gene therapy, which was accompanied by AMP-activated protein kinase (AMPK) phosphorylation and heme oxygenase (HO)-1 induction. Furthermore, AMPK-mediated peroxisome proliferator-activated receptor-α (PPARα) activation by APN was ascribable to HO-1 induction. Additionally, we revealed direct transcriptional regulation of HO-1 by the binding of PPARα to a PPAR-responsive element (PPRE) by various experimental assessments. Interestingly, overexpression of HO-1 in hepatocytes mimicked the protective effect of APN in attenuating iron-mediated injury, whereas it was abolished by SnPP and small interfering HO-1. Furthermore, bilirubin, the end-product of the HO-1 reaction, but not CO, protected hepatocytes from iron dextran-mediated caspase activation. Herein, we demonstrate a novel functional PPRE in the promoter regions of HO-1, and APN-mediated HO-1 induction elicited an antiapoptotic effect and a decrease in iron deposition in hepatocytes subjected to iron challenge.
Collapse
Affiliation(s)
- Heng Lin
- Institute of Pharmacology and Toxicology, Tzu-Chi University, Hualien, Taiwan
| | | | | | | | | | | | | |
Collapse
|
149
|
Taha H, Skrzypek K, Guevara I, Nigisch A, Mustafa S, Grochot-Przeczek A, Ferdek P, Was H, Kotlinowski J, Kozakowska M, Balcerczyk A, Muchova L, Vitek L, Weigel G, Dulak J, Jozkowicz A. Role of heme oxygenase-1 in human endothelial cells: lesson from the promoter allelic variants. Arterioscler Thromb Vasc Biol 2010; 30:1634-41. [PMID: 20508205 PMCID: PMC2906705 DOI: 10.1161/atvbaha.110.207316] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Heme oxygenase-1 (HO-1) is an antioxidative, antiinflammatory, and cytoprotective enzyme that is induced in response to cellular stress. The HO-1 promoter contains a (GT)n microsatellite DNA, and the number of GT repeats can influence the occurrence of cardiovascular diseases. We elucidated the effect of this polymorphism on endothelial cells isolated from newborns of different genotypes. METHODS AND RESULTS On the basis of HO-1 expression, we classified the HO-1 promoter alleles into 3 groups: short (S) (most active, GT < or = 23), medium (moderately active, GT=24 to 28), and long (least active, GT > or = 29). The presence of the S allele led to higher basal HO-1 expression and stronger induction in response to cobalt protoporphyrin, prostaglandin-J(2), hydrogen peroxide, and lipopolysaccharide. Cells carrying the S allele survived better under oxidative stress, a fact associated with the lower concentration of oxidized glutathione and more favorable oxidative status, as determined by measurement of the ratio of glutathione to oxidized glutathione. Moreover, they proliferated more efficiently in response to vascular endothelial growth factor A, although the vascular endothelial growth factor-induced migration and sprouting of capillaries were not influenced. Finally, the presence of the S allele was associated with lower production of some proinflammatory mediators, such as interleukin-1beta, interleukin-6, and soluble intercellular adhesion molecule-1. CONCLUSIONS The (GT)n promoter polymorphism significantly modulates a cytoprotective, proangiogenic, and antiinflammatory function of HO-1 in human endothelium.
Collapse
Affiliation(s)
- Hevidar Taha
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Klaudia Skrzypek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Ibeth Guevara
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | | | | | - Anna Grochot-Przeczek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Pawel Ferdek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Halina Was
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Jerzy Kotlinowski
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Magdalena Kozakowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | | | | | - Libor Vitek
- Charles University in Prague, Czech Republic
| | | | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| |
Collapse
|
150
|
Chen HH, Chen TW, Lin H. Pravastatin attenuates carboplatin-induced nephrotoxicity in rodents via peroxisome proliferator-activated receptor alpha-regulated heme oxygenase-1. Mol Pharmacol 2010; 78:36-45. [PMID: 20368269 DOI: 10.1124/mol.109.061101] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
The aim of this study was to explore the molecular mechanisms underlying the protective effect of pravastatin against carboplatin-induced nephrotoxicity in rodents. We exposed rat NRK-52E renal tubular epithelial cells to carboplatin, with or without pravastatin. Pravastatin decreased production of reactive oxygen species, increased expression of heme oxygenase-1 (HO-1), cyclooxygenase-2, and 6-keto prostaglandin F1alpha, enhanced nuclear translocation of peroxisome proliferator-activated receptor-alpha (PPARalpha), and increased HO-1 promoter and peroxisome proliferator response element (PPRE) activities. We found interaction of PPARalpha with PPRE on the HO-1 promoter in nuclear extracts from pravastatin-treated NRK-52E cells and by chromatin immunoprecipitation. We pretreated mice with pravastatin and then administered a single intraperitoneal injection of carboplatin. Effects on renal function, morphology, apoptosis, and survival were assessed. In response to carboplatin injection, mice developed acute renal failure, with elevated activated caspase-3, increased apoptotic bodies, and decreased survival. Pretreatment with pravastatin significantly ameliorated renal dysfunction and apoptosis and improved renal morphology and survival. Injection of pravastatin also induced overexpression of PPARalpha and HO-1 in wild-type mice, and HO-1 expression was significantly attenuated in PPARalpha-knockout mice. These results indicate that pravastatin up-regulates HO-1 and protects against carboplatin-induced renal dysfunction and apoptosis via a PPARalpha-dependent pathway.
Collapse
Affiliation(s)
- Hsi-Hsien Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | | | | |
Collapse
|