101
|
Tousoulis D, Simopoulou C, Papageorgiou N, Oikonomou E, Hatzis G, Siasos G, Tsiamis E, Stefanadis C. Endothelial dysfunction in conduit arteries and in microcirculation. Novel therapeutic approaches. Pharmacol Ther 2014; 144:253-267. [PMID: 24928320 DOI: 10.1016/j.pharmthera.2014.06.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 05/28/2014] [Indexed: 11/22/2022]
Abstract
The vascular endothelium not only is a single monolayer of cells between the vessel lumen and the intimal wall, but also plays an important role by controlling vascular function and structure mainly via the production of nitric oxide (NO). The so called "cardiovascular risk factors" are associated with endothelial dysfunction, that reduces NO bioavailability, increases oxidative stress, and promotes inflammation contributing therefore to the development of atherosclerosis. The significant role of endothelial dysfunction in the development of atherosclerosis emphasizes the need for efficient therapeutic interventions. During the last years statins, angiotensin-converting enzyme inhibitors, angiotensin-receptor antagonists, antioxidants, beta-blockers and insulin sensitizers have been evaluated for their ability to restore endothelial function (Briasoulis et al., 2012). As there is not a straightforward relationship between therapeutic interventions and improvement of endothelial function but rather a complicated interrelationship between multiple cellular and sub-cellular targets, research has been focused on the understanding of the underlying mechanisms. Moreover, the development of novel diagnostic invasive and non-invasive methods has allowed the early detection of endothelial dysfunction expanding the role of therapeutic interventions and our knowledge. In the current review we present the available data concerning the contribution of endothelial dysfunction to atherogenesis and review the methods that assess endothelial function with a view to understand the multiple targets of therapeutic interventions. Finally we focus on the classic and novel therapeutic approaches aiming to improve endothelial dysfunction and the underlying mechanisms.
Collapse
Affiliation(s)
- Dimitris Tousoulis
- 1st Cardiology Department, Athens University Medical School, Hippokration Hospital, Greece.
| | - Chryssa Simopoulou
- 1st Cardiology Department, Athens University Medical School, Hippokration Hospital, Greece
| | - Nikos Papageorgiou
- 1st Cardiology Department, Athens University Medical School, Hippokration Hospital, Greece
| | - Evangelos Oikonomou
- 1st Cardiology Department, Athens University Medical School, Hippokration Hospital, Greece
| | - George Hatzis
- 1st Cardiology Department, Athens University Medical School, Hippokration Hospital, Greece
| | - Gerasimos Siasos
- 1st Cardiology Department, Athens University Medical School, Hippokration Hospital, Greece
| | - Eleftherios Tsiamis
- 1st Cardiology Department, Athens University Medical School, Hippokration Hospital, Greece
| | | |
Collapse
|
102
|
Sertznig P, Reichrath J. Peroxisome proliferator-activated receptors (PPARs) in dermatology. DERMATO-ENDOCRINOLOGY 2014. [DOI: 10.4161/derm.15025] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
103
|
El-Bassossy HM, Abo-Warda SM, Fahmy A. Chrysin and luteolin alleviate vascular complications associated with insulin resistance mainly through PPAR-γ activation. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2014; 42:1153-67. [PMID: 25169908 DOI: 10.1142/s0192415x14500724] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Chrysin and luteolin are two flavonoids with Peroxisome proliferators-activated receptor γ (PPAR-γ) stimulating activity. Here, we investigated the protective effect of chrysin and luteolin from vascular complications associated with insulin resistance (IR). IR was induced in rats by drinking fructose for 12 weeks while chrysin and luteolin were given for 6 weeks with or without PPAR-γ antagonist, bisphenol A diglycidyl ether (BADGE). Then, blood pressure (BP) was recorded and serum levels of glucose, insulin, advanced glycation end products (AGEs) and lipids were measured. Concentration response curves for phenylephrine (PE), KCl, and acetylcholine (ACh) were obtained in thoracic aorta rings. Aortic reactive oxygen species (ROS) and nitric oxide (NO) generation were also studied. Chrysin and luteolin significantly alleviated systolic BP elevations caused by IR, while the co-administration of BADGE prevented chrysin alleviation. Although, neither chrysin nor luteolin affected ACh impaired vasodilatation, they both alleviated exaggerated vasoconstrictions to PE and KCl in IR animals. In addition, incubation of the aorta from IR animals with chrysin or luteolin prevented exaggerated vasoconstrictions to PE and KCl. On the other hand, co-administration of BADGE or co-incubation with GW9662, the selective PPAR-γ antagonist, prevented chrysin alleviation. Both chrysin and luteolin inhibited the developed hyperinsulinemia and increases in serum AGEs, lipids while, BADGE reduced the effect of chrysin on hyperinsulinemia and dyslipidemia. Chrysin and luteolin markedly inhibited elevated NO and ROS in IR aortae while BADGE did not change their effect on NO and ROS. In conclusion, chrysin and luteolin alleviate vascular complications associated with IR mainly through PPAR-γ dependent pathways.
Collapse
Affiliation(s)
- Hany M El-Bassossy
- Department of Pharmacology, Faculty of Pharmacy, King Abdulaziz University, Kingdom of Saudi Arabia , Department of Pharmacology, Faculty of Pharmacy, Zagazig University, Egypt
| | | | | |
Collapse
|
104
|
Mineralocorticoid receptor: a critical player in vascular remodeling. SCIENCE CHINA-LIFE SCIENCES 2014; 57:809-17. [PMID: 25104454 DOI: 10.1007/s11427-014-4691-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 06/06/2014] [Indexed: 01/10/2023]
Abstract
Vascular remodeling is a pathological condition with structural changes of blood vessels. Both inside-out and outside-in hypothesis have been put forward to describe mechanisms of vascular remodeling. An integrated model of these two hypotheses emphasizes the importance of immune cells such as monocytes/macrophages, T cells, and dendritic cells. These immune cells are at the center stage to orchestrate cellular proliferation, migration, and interactions of themselves and other vascular cells including endothelial cells (ECs), vascular smooth muscle cells (VSMCs), and fibroblasts. These changes on vascular wall lead to inflammation and oxidative stress that are largely responsible for vascular remodeling. Mineralocorticoid receptor (MR) is a classic nuclear receptor. MR agonist promotes inflammation and oxidative stress and therefore exacerbates vascular remodeling. Conversely, MR antagonists have the opposite effects. MR has direct roles on vascular cells through non-genomic or genomic actions to modulate inflammation and oxidative stress. Recent studies using genetic mouse models have revealed that MR in myeloid cells, VSMCs and ECs all contribute to vascular remodeling. In conclusion, data in the past years have demonstrated that MR is a critical control point in modulating vascular remodeling. Studies will continue to provide evidence with more detailed mechanisms to support this notion.
Collapse
|
105
|
Zhang SM, Zhu LH, Li ZZ, Wang PX, Chen HZ, Guan HJ, Jiang DS, Chen K, Zhang XF, Tian S, Yang D, Zhang XD, Li H. Interferon regulatory factor 3 protects against adverse neo-intima formation. Cardiovasc Res 2014; 102:469-479. [PMID: 24596398 DOI: 10.1093/cvr/cvu052] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/16/2024] Open
Abstract
AIMS Vascular smooth muscle cell (VSMC) proliferation is central to the pathophysiology of neo-intima formation. Interferon regulatory factor 3 (IRF3) inhibits the growth of cancer cells and fibroblasts. However, the role of IRF3 in vascular neo-intima formation is unknown. We evaluated the protective role of IRF3 against neo-intima formation in mice and the underlying mechanisms. METHODS AND RESULTS IRF3 expression was down-regulated in VSMCs after carotid wire injury in vivo, and in SMCs after platelet-derived growth factor (PDGF)-BB challenge in vitro. Global knockout of IRF3 (IRF3-KO) led to accelerated neo-intima formation and proliferation of VSMCs, whereas the opposite was seen in SMC-specific IRF3 transgenic mice. Mechanistically, we identified IRF3 as a novel regulator of peroxisome proliferator-activated receptor γ (PPARγ), a negative regulator of SMC proliferation after vascular injury. Binding of IRF3 to the AB domain of PPARγ in the nucleus of SMCs facilitated PPARγ transactivation, resulting in decreased proliferation cell nuclear antigen expression and suppressed proliferation. Overexpression of wild-type, but not truncated, IRF3 with a mutated IRF association domain (IAD) retained the ability to exert anti-proliferative effect. CONCLUSIONS IRF3 inhibits VSMC proliferation and neo-intima formation after vascular injury through PPARγ activation.
Collapse
Affiliation(s)
- Shu-Min Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Jiefang Road 238, Wuhan 430060, China Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Li-Hua Zhu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Jiefang Road 238, Wuhan 430060, China Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Zuo-Zhi Li
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pi-Xiao Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Jiefang Road 238, Wuhan 430060, China Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Hou-Zao Chen
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hong-Jing Guan
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Jiefang Road 238, Wuhan 430060, China Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Ding-Sheng Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Jiefang Road 238, Wuhan 430060, China Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Ke Chen
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Xiao-Fei Zhang
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Song Tian
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Jiefang Road 238, Wuhan 430060, China Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Da Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Jiefang Road 238, Wuhan 430060, China Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Xiao-Dong Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Jiefang Road 238, Wuhan 430060, China Cardiovascular Research Institute of Wuhan University, Wuhan, China
| |
Collapse
|
106
|
Pentraxin 3 promotes oxLDL uptake and inhibits cholesterol efflux from macrophage-derived foam cells. Exp Mol Pathol 2014; 96:292-9. [DOI: 10.1016/j.yexmp.2014.03.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Revised: 03/13/2014] [Accepted: 03/17/2014] [Indexed: 11/17/2022]
|
107
|
Zhao R, Feng J, He G. miR-613 regulates cholesterol efflux by targeting LXRα and ABCA1 in PPARγ activated THP-1 macrophages. Biochem Biophys Res Commun 2014; 448:329-34. [DOI: 10.1016/j.bbrc.2014.04.052] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 04/10/2014] [Indexed: 11/25/2022]
|
108
|
Liu J, Zhuo XZ, Liu W, Wan Z, Wang X, Zhang W, Gao S, Yuan Z, Wu Y. Drug-Eluting Stent, but Not Bare Metal Stent, Accentuates the Systematic Inflammatory Response in Patients. Cardiology 2014; 128:259-65. [DOI: 10.1159/000358508] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 01/09/2014] [Indexed: 11/19/2022]
|
109
|
Abstract
The clinical recognition of pulmonary arterial hypertension (PAH) is increasing, and with recent therapeutic advances, short-term survival has improved. In spite of these advances, however, PAH remains a disease with substantial morbidity and long-term mortality. The pathogenesis of PAH involves a complex interaction of local and distant cytokines, growth factors, co-factors, and transcription factors occurring in the right genetic and environmental setting. These factors ultimately lead to the detrimental changes in vascular anatomy and function seen in PAH patients. An important association between obesity/insulin resistance and PAH has recently been identified. Both conditions occur in the presence of a chronic low-grade inflammatory state, and although it is unlikely that a single pathway is solely responsible for the observed association, deficiencies in adiponectin, apolipoprotein E (ApoE) and peroxisome proliferator-activator receptor gamma (PPAR-γ) activity likely play a prominent role. Although incompletely understood, it is clear that further investigation is warranted and the role of weight loss and improved glycemic control in the treatment of at-risk patients with PAH and obesity should be determined.
Collapse
Affiliation(s)
- Elisa A Bradley
- Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center and Nationwide Children's Hospital, Columbus, OH, USA
| | - David Bradley
- Division of Endocrinology, Diabetes and Metabolism, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
110
|
Reciprocal modulation of surface expression of annexin A2 in a human umbilical vein endothelial cell-derived cell line by eicosapentaenoic acid and docosahexaenoic acid. PLoS One 2014; 9:e85045. [PMID: 24465474 PMCID: PMC3897403 DOI: 10.1371/journal.pone.0085045] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 11/25/2013] [Indexed: 11/20/2022] Open
Abstract
Background Annexin A2 (ANXA2), a member of the annexin family of cytosolic Ca2+-binding proteins, plays a pivotal role in vascular biology. Small amounts of this protein and S100A10 protein are exposed on the surface of endothelial cells (ECs). They control fibrinolysis by recruiting tissue-type and urokinase-type plasminogen activators from the plasma. Nutritional studies indicate that two major long-chain polyunsaturated fatty acids (PUFAs), i.e., eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), provide benefits for EC functions. The effects of EPA and DHA on the plasminogen/plasmin system have not been characterized. Methodology/Principal Findings Proteomic analysis of a cultured human umbilical vein EC-derived cell line, HUV-EC-C, showed that cell-associated ANXA2 decreased with EPA treatment and increased with DHA. A small fraction of ANXA2 was bound to the cell surface, which was also affected by these PUFAs following the same trends. Cell surface expression was negatively regulated by protein kinase C (PKC) α-mediated Ser-phosphorylation, which was up- and down-regulated by EPA and DHA, respectively. These PUFAs differentially affected a small fraction of caveolae/rafts-associated ANXA2. In addition to chymotrypsin-like activity in the serum, newly activated plasmin cleaved the ANXA2 on the cell surface at distinct sites in the N-terminal sequence. ANXA2 also bound to membranes released in the medium, which was similarly processed by these proteases. Both the PUFAs did not directly affect the release. Conclusion/Significance These results suggest that EPA and DHA reciprocally control cell surface location of ANXA2. Moreover, cleavage of this protein by plasmin likely resulted in autodigestion of the platform for formation of this protease. In conjunction with termination of the proteolysis by rapid inactivation of plasmin by α-2-antiplasmin and other polypeptide inhibitors, this feedback mechanism may emphasize the benefits of these PUFA in regulation of the initiation of fibrinolysis on the surface of ECs.
Collapse
|
111
|
Wan J, Xiao Z, Chao S, Xiong S, Gan X, Qiu X, Xu C, Ma Y, Tu X. Pioglitazone modulates the proliferation and apoptosis of vascular smooth muscle cells via peroxisome proliferators-activated receptor-gamma. Diabetol Metab Syndr 2014; 6:101. [PMID: 25302079 PMCID: PMC4190377 DOI: 10.1186/1758-5996-6-101] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 08/10/2014] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND PPARγ is a member of the nuclear hormone receptor superfamily. It has been considered as a mediator regulating metabolism, anti-inflammation, and pro-proliferation in the Vascular Smooth Muscle Cells (VSMCs). Thiazolidinediones (TZDs), synthetic ligands of PPARγ, have anti-proliferative and pro-apoptotic effects on VSMCs, which prevent the formation and progression of atherosclerosis and restenosis following percutaneous coronary intervention (PCI). However, the underlying mechanism remains elusive. This present study therefore aimed to investigate the signaling pathway by which pioglitazone, one of TZDs, inhibits proliferation and induces apoptosis of VSMCs. METHODS The effects of pioglitazone on VSMC proliferation and apoptosis were studied. Cell proliferation was determined using BrdU incorporation assay. Cell apoptosis was monitored with Hoechst and Annexin V staining. The expression of caspases and cyclins was determined using real-time PCR and Western blot. RESULTS Pioglitazone treatment and PPARγ overexpression inhibited proliferation and induced apoptosis of VSMCs, whereas blocking by antagonist or silencing by siRNA of PPARγ significantly attenuated pioglitazone's effect. Furthermore, pioglitazone treatment or PPARγ overexpression increased caspase 3 and caspase 9 expression, and decreased the expression of cyclin B1 and cyclin D1 in VSMCs. CONCLUSIONS Pioglitazone inhibits VSMCs proliferation and promotes apoptosis of VSMCs through a PPARγ signaling pathway. Up-regulation of caspase 3 and down-regulation of cyclins mediates pioglitazone's anti-proliferative and pro-apoptotic effects. Our results imply that pioglitazone prevents the VSMCs proliferation via modulation of caspase and cyclin signaling pathways in a PPARγ-dependent manner.
Collapse
Affiliation(s)
- Jing Wan
- />Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei China
| | - Zhichao Xiao
- />Department of Cardiology, Tongji Medical College of Huazhong University of Science and Technology affiliated Tongji Hospital, Wuhan, Hubei China
| | - Shengping Chao
- />Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei China
| | - Shixi Xiong
- />Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei China
| | - Xuedong Gan
- />Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei China
| | - Xuguang Qiu
- />Department of Cardiology, Tongji Medical College of Huazhong University of Science and Technology affiliated Tongji Hospital, Wuhan, Hubei China
| | - Chang Xu
- />Department of Cardiology, Tongji Medical College of Huazhong University of Science and Technology affiliated Tongji Hospital, Wuhan, Hubei China
| | - Yexin Ma
- />Department of Cardiology, Tongji Medical College of Huazhong University of Science and Technology affiliated Tongji Hospital, Wuhan, Hubei China
| | - Xin Tu
- />Cardiovascular Research, Life Science and Technology College, Human Genome Research Center, Huazhong University of Science and Technology, Wuhan, Hubei China
| |
Collapse
|
112
|
Mansour M. The Roles of Peroxisome Proliferator-Activated Receptors in the Metabolic Syndrome. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 121:217-66. [DOI: 10.1016/b978-0-12-800101-1.00007-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
113
|
Nehrenheim K, Meyer I, Brenden H, Vielhaber G, Krutmann J, Grether-Beck S. Dihydrodehydrodiisoeugenol enhances adipocyte differentiation and decreases lipolysis in murine and human cells. Exp Dermatol 2013; 22:638-43. [DOI: 10.1111/exd.12218] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2013] [Indexed: 12/17/2022]
Affiliation(s)
- Katja Nehrenheim
- IUF - Leibniz Research Institute for Environmental Medicine; Düsseldorf; Germany
| | | | - Heidi Brenden
- IUF - Leibniz Research Institute for Environmental Medicine; Düsseldorf; Germany
| | | | - Jean Krutmann
- IUF - Leibniz Research Institute for Environmental Medicine; Düsseldorf; Germany
| | - Susanne Grether-Beck
- IUF - Leibniz Research Institute for Environmental Medicine; Düsseldorf; Germany
| |
Collapse
|
114
|
Trombetta A, Togliatto G, Rosso A, Dentelli P, Olgasi C, Cotogni P, Brizzi MF. Increase of palmitic acid concentration impairs endothelial progenitor cell and bone marrow-derived progenitor cell bioavailability: role of the STAT5/PPARγ transcriptional complex. Diabetes 2013; 62:1245-57. [PMID: 23223023 PMCID: PMC3609587 DOI: 10.2337/db12-0646] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Metabolic profiling of plasma nonesterified fatty acids discovered that palmitic acid (PA), a natural peroxisome proliferator-activated receptor γ (PPARγ) ligand, is a reliable type 2 diabetes biomarker. We investigated whether and how PA diabetic (d-PA) concentrations affected endothelial progenitor cell (EPC) and bone marrow-derived hematopoietic cell (BM-HC) biology. PA physiologic (n-PA) and d-PA concentrations were used. Proliferating cell nuclear antigen content and signal transducer and activator of transcription 5 (STAT5), PPARγ, cyclin D1, and p21(Waf) expression were evaluated. Small interfering RNA technology, gene reporter luciferase assay, electrophoretic mobility shift assay, chromatin immunoprecipitation assay, and coimmunoprecipitation were exploited. In vivo studies and migration assays were also performed. d-PA, unlike n-PA or physiological and diabetic oleic and stearic acid concentrations, impaired EPC migration and EPC/BM-HC proliferation through a PPARγ-mediated STAT5 transcription inhibition. This event did not prevent the formation of a STAT5/PPARγ transcriptional complex but was crucial for gene targeting, as p21(Waf) gene promoter, unlike cyclin D1, was the STAT5/PPARγ transcriptional target. Similar molecular events could be detected in EPCs isolated from type 2 diabetic patients. By expressing a constitutively activated STAT5 form, we demonstrated that STAT5 content is crucial for gene targeting and EPC fate. Finally, we also provide in vivo data that d-PA-mediated EPC dysfunction could be rescued by PPARγ blockade. These data provide first insights on how mechanistically d-PA drives EPC/BM-HC dysfunction in diabetes.
Collapse
Affiliation(s)
| | | | - Arturo Rosso
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Cristina Olgasi
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Paolo Cotogni
- Department of Anesthesiology and Intensive Care, University of Turin, Turin, Italy
| | - Maria Felice Brizzi
- Department of Medical Sciences, University of Turin, Turin, Italy
- Corresponding author: Maria Felice Brizzi,
| |
Collapse
|
115
|
Wang Y, Yu L, Cui N, Jin X, Zhu D, Jiang C. Differential sensitivities of the vascular K(ATP) channel to various PPAR activators. Biochem Pharmacol 2013; 85:1495-503. [PMID: 23500542 DOI: 10.1016/j.bcp.2013.02.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 02/26/2013] [Accepted: 02/27/2013] [Indexed: 12/11/2022]
Abstract
Several agonists of the peroxisome proliferator-activated receptors (PPARs) are currently used for the treatment of metabolic disorders including diabetes. We have recently shown that one of them, Rosiglitazone, inhibits the vascular ATP-sensitive K⁺ (K(ATP)) channel and compromises the coronary vasodilation by the β-adrenoceptor agonist. Here, we show evidence for the channel inhibition by various PPAR agonists, information that may be useful for finding new therapeutical agents with less cardiovascular side-effects and more selective K(ATP) channel blockers targeting at the K(ir)6.1 subunit. Structural comparison of these PPAR agonists may shed insight into the critical chemical groups for the channel inhibition. K(ir)6.1/SUR2B channel was expressed in HEK293 cells and studied in whole-cell voltage clamp. The K(ir)6.1/SUR2B channel was strongly inhibited by several PPAR(γ) agonists with potencies similar to, or higher than, that of Rosiglitazone, while other PPAR(γ) agonists barely inhibited the channel. The K(ir)6.1/SUR2B channel was also inhibited by PPAR(α) and PPAR(β/δ) agonists with intermediate potencies. The structure necessary for the channel inhibition appears to include the thiazole linked to an aromatic or furan ring. Additions of side groups such as small aliphatic chain increased the potency for channel inhibition, while additions of aromatic rings reduced it. These results indicate that the PPAR(γ) agonists with weak K(ATP) channel inhibition may be potential candidates as therapeutical agents, and those with strong channel inhibition may be used as selective K(ATP) channel blockers. The structural information of the PPAR agonists may be useful for the development of new therapeutical modalities with less cardiovascular side-effects.
Collapse
Affiliation(s)
- Yingji Wang
- Department of Biology, Georgia State University, Atlanta, GA 30302-4010, USA
| | | | | | | | | | | |
Collapse
|
116
|
Valero-Muñoz M, Martín-Fernández B, Ballesteros S, Martínez-Martínez E, Blanco-Rivero J, Balfagón G, Cachofeiro V, Lahera V, de las Heras N. Relevance of vascular peroxisome proliferator-activated receptor γ coactivator-1α to molecular alterations in atherosclerosis. Exp Physiol 2013; 98:999-1008. [DOI: 10.1113/expphysiol.2012.070557] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
117
|
Auclair M, Vigouroux C, Boccara F, Capel E, Vigeral C, Guerci B, Lascols O, Capeau J, Caron-Debarle M. Peroxisome proliferator-activated receptor-γ mutations responsible for lipodystrophy with severe hypertension activate the cellular renin-angiotensin system. Arterioscler Thromb Vasc Biol 2013; 33:829-38. [PMID: 23393388 DOI: 10.1161/atvbaha.112.300962] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Inactivating peroxisome proliferator-activated receptor-γ (PPARγ) mutations lead to a syndrome of familial partial lipodystrophy (FPLD3) associated with early-onset severe hypertension. PPARγ can repress the vascular renin-angiotensin system (RAS) and angiotensin II receptor 1 expression. We evaluated the relationships between PPARγ inactivation and cellular RAS using FPLD3 patients' cells and human vascular smooth muscle cells expressing mutant or wild-type PPARγ. Approach and Results- We identified 2 novel PPARG mutations, R165T and L339X, located in the DNA and ligand-binding domains of PPARγ, respectively in 4 patients from 2 FPLD3 families. In cultured skin fibroblasts and peripheral blood mononuclear cells from the 4 patients and healthy controls, we compared markers of RAS activation, oxidative stress, and inflammation, and tested the effect of modulators of PPARγ and angiotensin II receptor 1. We studied the impact of the 2 mutations on the transcriptional activity of PPARγ and on the vascular RAS in transfected human vascular smooth muscle cells. Systemic RAS was not altered in patients. However, RAS markers were overexpressed in patients' fibroblasts and peripheral blood mononuclear cells, as in vascular cells expressing mutant PPARγ. Angiotensin II-mediated mitogen-activated protein kinase activity increased in patients' fibroblasts, consistent with RAS constitutive activation. Patients' cells also displayed oxidative stress and inflammation. PPARγ activation and angiotensin II receptor 1 mRNA silencing reversed RAS overactivation, oxidative stress, and inflammation, arguing for a role of angiotensin II receptor 1 in these processes. CONCLUSIONS Two novel FPLD3-linked PPARG mutations are associated with a defective transrepression of cellular RAS leading to cellular dysfunction, which might contribute to the specific FPLD3-linked severe hypertension.
Collapse
Affiliation(s)
- Martine Auclair
- INSERM UMRS938, Centre de Recherche Saint Antoine, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
118
|
Kathuria S, Mahadevan N, Balakumar P. Possible involvement of PPARγ-associated eNOS signaling activation in rosuvastatin-mediated prevention of nicotine-induced experimental vascular endothelial abnormalities. Mol Cell Biochem 2013; 374:61-72. [PMID: 23149826 DOI: 10.1007/s11010-012-1505-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 10/25/2012] [Indexed: 12/30/2022]
Abstract
Nicotine exposure via cigarette smoking and tobacco chewing is associated with vascular complications. The present study investigated the effect of rosuvastatin in nicotine (2 mg/kg/day, i.p., 4 weeks)-induced vascular endothelial dysfunction (VED) in rats. The development of VED was assessed by employing isolated aortic ring preparation and estimating aortic and serum nitrite/nitrate concentration. Further, scanning electron microscopy and hematoxylin-eosin staining of thoracic aorta were performed to assess the vascular endothelial integrity. Moreover, oxidative stress was assessed by estimating aortic superoxide anion generation and serum thiobarbituric acid-reactive substances. The nicotine administration produced VED by markedly reducing acetylcholine-induced endothelium-dependent relaxation, impairing the integrity of vascular endothelium, decreasing aortic and serum nitrite/nitrate concentration, increasing oxidative stress, and inducing lipid alteration. However, treatment with rosuvastatin (10 mg/kg/day, i.p., 4 weeks) markedly attenuated nicotine-induced vascular endothelial abnormalities, oxidative stress, and lipid alteration. Interestingly, the co-administration of peroxisome proliferator-activated receptor γ (PPARγ) antagonist, GW9662 (1 mg/kg/day, i.p., 2 weeks) submaximally, significantly prevented rosuvastatin-induced improvement in vascular endothelial integrity, endothelium-dependent relaxation, and nitrite/nitrate concentration in rats administered nicotine. However, GW9662 co-administration did not affect rosuvastatin-associated vascular anti-oxidant and lipid-lowering effects. The incubation of aortic ring, isolated from rosuvastatin-treated nicotine-administered rats, with L-NAME (100 μM), an inhibitor of nitric oxide synthase (NOS), significantly attenuated rosuvastatin-induced improvement in acetylcholine-induced endothelium-dependent relaxation. Rosuvastatin prevents nicotine-induced vascular endothelial abnormalities by activating PPARγ and endothelial NOS signaling pathways. Moreover, the PPARγ-independent anti-oxidant and lipid-lowering effects of rosuvastatin might additionally play a role in the improvement of vascular endothelial function.
Collapse
Affiliation(s)
- Sonam Kathuria
- Cardiovascular Pharmacology Division, Department of Pharmacology, Rajendra Institute of Technology and Sciences, Sirsa, 125 055, India
| | | | | |
Collapse
|
119
|
Tiyerili V, Becher UM, Aksoy A, Lütjohann D, Wassmann S, Nickenig G, Mueller CFH. AT1-receptor-deficiency induced atheroprotection in diabetic mice is partially mediated via PPARγ. Cardiovasc Diabetol 2013; 12:30. [PMID: 23374104 PMCID: PMC3667017 DOI: 10.1186/1475-2840-12-30] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2012] [Accepted: 01/28/2013] [Indexed: 11/24/2022] Open
Abstract
Objective Peroxisome-proliferator–activated-receptor-γ (PPARγ) acts as a transcriptional regulator of multiple genes involved in glucose and lipid metabolism. In vitro studies showed that activated PPARγ suppresses AT1R-gene expression and vice versa. However, it has not yet been determined in vivo, whether AT1R-PPARγ-interactions play a relevant role in the pathogenesis of diabetic complications and specifically in accelerated atherosclerosis. Methods and results ApoE−/− and ApoE−/−/AT1R−/−-mice were rendered diabetic by intraperitoneal injections of streptozotocin. Diabetic and non-diabetic ApoE−/−-mice were further randomized to receive the AT1R antagonist telmisartan, the selective PPARγ antagonist GW9662, telmisartan and GW9662 or vehicle for 18 weeks. Diabetic and non-diabetic ApoE−/−/AT1R−/−-mice were randomized to receive either GW9662 or vehicle. GW9662 treatment in diabetic ApoE−/− and diabetic ApoE−/−/AT1−/−-mice resulted in the highest elevation of fasting blood glucose levels, whereas telmisartan treatment and AT1 deficiency in ApoE−/−-mice showed the lowest fasting blood glucose levels. Diabetic ApoE−/−-mice displayed severe impairment of endothelial function, enhanced oxidative stress and increased atherosclerotic lesion formation. ApoE−/−/AT1R−/− and telmisartan-treated ApoE−/−-mice showed a significantly better endothelial function, decreased oxidative stress and reduced atherosclerotic lesion formation. Treatment of diabetic ApoE−/− and ApoE−/−/AT1R−/−-mice with the selective PPARγ antagonist GW9662 omitted the atheroprotective effects of AT1R deficiency or AT1 antagonism. Conclusion Genetic disruption or pharmacological inhibition of the AT1R attenuates atherosclerosis and improves endothelial function in diabetic ApoE−/−-mice via the PPARγ pathway.
Collapse
Affiliation(s)
- Vedat Tiyerili
- Medizinische Klinik und Poliklinik II, Innere Medizin, Universitätsklinikum Bonn, Sigmund Freud Str, 25, 53105, Bonn, Germany.
| | | | | | | | | | | | | |
Collapse
|
120
|
Goto K, Iso T, Hanaoka H, Yamaguchi A, Suga T, Hattori A, Irie Y, Shinagawa Y, Matsui H, Syamsunarno MRAA, Matsui M, Haque A, Arai M, Kunimoto F, Yokoyama T, Endo K, Gonzalez FJ, Kurabayashi M. Peroxisome proliferator-activated receptor-γ in capillary endothelia promotes fatty acid uptake by heart during long-term fasting. J Am Heart Assoc 2013; 2:e004861. [PMID: 23525438 PMCID: PMC3603264 DOI: 10.1161/jaha.112.004861] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background Endothelium is a crucial blood–tissue interface controlling energy supply according to organ needs. We investigated whether peroxisome proliferator‐activated receptor‐γ (PPARγ) induces expression of fatty acid–binding protein 4 (FABP4) and fatty acid translocase (FAT)/CD36 in capillary endothelial cells (ECs) to promote FA transport into the heart. Methods and Results Expression of FABP4 and CD36 was induced by the PPARγ agonist pioglitazone in human cardiac microvessel ECs (HCMECs), but not in human umbilical vein ECs. Real‐time PCR and immunohistochemistry of the heart tissue of control (Ppargfl/null) mice showed an increase in expression of FABP4 and CD36 in capillary ECs by either pioglitazone treatment or 48 hours of fasting, and these effects were not found in mice deficient in endothelial PPARγ (Pparg∆EC/null). Luciferase reporter constructs of the Fabp4 and CD36 promoters were markedly activated by pioglitazone in HCMECs through canonical PPAR‐responsive elements. Activation of PPARγ facilitated FA uptake by HCMECs, which was partially inhibited by knockdown of either FABP4 or CD36. Uptake of an FA analogue, 125I‐BMIPP, was significantly reduced in heart, red skeletal muscle, and adipose tissue in Pparg∆EC/null mice as compared with Ppargfl/null mice after olive oil loading, whereas those values were comparable between Ppargfl/null and Pparg∆EC/null null mice on standard chow and a high‐fat diet. Furthermore, Pparg∆EC/null mice displayed slower triglyceride clearance after olive oil loading. Conclusions These findings identified a novel role for capillary endothelial PPARγ as a regulator of FA handing in FA‐metabolizing organs including the heart in the postprandial state after long‐term fasting.
Collapse
Affiliation(s)
- Kosaku Goto
- Department of Medicine and Biological Science, Gunma University Graduate School of Health Sciences, Maebashi, Gunma, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
121
|
Salama RM, Schaalan MF, Ibrahim ME, Khalifa AE, Elkoussi AA. Effectiveness of Telmisartan as an Adjunct to Metformin in Treating Type II Diabetes Mellitus in Rats. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/ojemd.2013.33026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
122
|
Balakumar P, Kathuria S. Submaximal PPARγ activation and endothelial dysfunction: new perspectives for the management of cardiovascular disorders. Br J Pharmacol 2012; 166:1981-92. [PMID: 22404217 DOI: 10.1111/j.1476-5381.2012.01938.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
PPARγ activation plays an important role in glucose metabolism by enhancing insulin sensitization. PPARγ is a primary target for thiazolidinedione-structured insulin sensitizers like pioglitazone and rosiglitazone employed for the treatment of type 2 diabetes mellitus. Additionally, PPARγ activation inhibits adhesion cascades and detrimental vascular inflammatory events. Importantly, activation of PPARγ plays a distinctive role in regulating the physiology and expression of endothelial nitric oxide synthase (eNOS) in the endothelium, resulting in enhanced generation of vascular nitric oxide. The PPARγ activation-mediated vascular anti-inflammatory and direct endothelial functional regulatory actions could, therefore, be beneficial in improving the vascular function in patients with atherosclerosis and hypertension with or without diabetes mellitus. Despite the disappointing cardiac side effect profile of rosiglitazone-like PPARγ full agonists, the therapeutic potential of novel pharmacological agents targeting PPARγ submaximally cannot be ruled out. This review discusses the potential regulatory role of PPARγ on eNOS expression and activation in improving the function of vascular endothelium. We argue that partial/submaximal activation of PPARγ could be a major target for vascular endothelial functional improvement. Interestingly, newly synthesized partial agonists of PPARγ such as balaglitazone, MBX-102, MK-0533, PAR-1622, PAM-1616, KR-62776 and SPPARγM5 are devoid of or have a reduced tendency to cause the adverse effects associated with full agonists of PPARγ. We propose that the vascular protective properties of pharmacological agents, which submaximally activate PPARγ, should be investigated. Moreover, the therapeutic opportunities of agents that submaximally activate PPARγ in preventing vascular endothelial dysfunction (VED) and VED-associated cardiovascular disorders are discussed.
Collapse
Affiliation(s)
- Pitchai Balakumar
- Cardiovascular Pharmacology Division, Department of Pharmacology, Institute of Pharmacy, Rajendra Institute of Technology and Sciences-RITS, Sirsa, India.
| | | |
Collapse
|
123
|
Spigoni V, Picconi A, Cito M, Ridolfi V, Bonomini S, Casali C, Zavaroni I, Gnudi L, Metra M, Dei Cas A. Pioglitazone improves in vitro viability and function of endothelial progenitor cells from individuals with impaired glucose tolerance. PLoS One 2012; 7:e48283. [PMID: 23139771 PMCID: PMC3489677 DOI: 10.1371/journal.pone.0048283] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 09/21/2012] [Indexed: 01/01/2023] Open
Abstract
Background Evidence suggests that the PPARγ-agonist insulin sensitizer pioglitazone, may provide potential beneficial cardiovascular (CV) effects beyond its anti-hyperglycaemic function. A reduced endothelial progenitor cell (EPC) number is associated with impaired glucose tolerance (IGT) or diabetes, conditions characterised by increased CV risk. Aim To evaluate whether pioglitazone can provide benefit in vitro in EPCs obtained from IGT subjects. Materials and Methods Early and late-outgrowth EPCs were obtained from peripheral blood mononuclear cells of 14 IGT subjects. The in vitro effect of pioglitazone (10 µM) with/without PPARγ-antagonist GW9662 (1 µM) was assessed on EPC viability, apoptosis, ability to form tubular-like structures and pro-inflammatory molecule expression. Results Pioglitazone increased early and late-outgrowth EPC viability, with negligible effects on apoptosis. The capacity of EPCs to form tubular-like structures was improved by pioglitazone in early (mean increase 28%; p = 0.005) and late-outgrowth (mean increase 30%; p = 0.037) EPCs. Pioglitazone reduced ICAM-1 and VCAM-1 adhesion molecule expression in both early (p = 0.001 and p = 0.012 respectively) and late-outgrowth (p = 0.047 and p = 0.048, respectively) EPCs. Similarly, pioglitazone reduced TNFα gene and protein expression in both early (p = 0.034;p = 0.022) and late-outgrowth (p = 0.026;p = 0.017) EPCs compared to control. These effects were prevented by incubation with the PPARγ-antagonist GW9662. Conclusion Pioglitazone exerts beneficial effects in vitro on EPCs isolated from IGT subjects, supporting the potential implication of pioglitazone as a CV protective agents.
Collapse
Affiliation(s)
- Valentina Spigoni
- Cardiology, Department of Experimental and Applied Medicine, University of Brescia, Brescia, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
124
|
Ohshima K, Mogi M, Nakaoka H, Jing F, Iwanami J, Min LJ, Tsukuda K, Kanno H, Ogimoto A, Higaki J, Horiuchi M. Inhibition of MCP-1/CCR2 signaling pathway is involved in synergistic inhibitory effects of irbesartan with rosuvastatin on vascular remodeling. ACTA ACUST UNITED AC 2012; 6:375-84. [PMID: 23107893 DOI: 10.1016/j.jash.2012.10.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 09/15/2012] [Accepted: 10/01/2012] [Indexed: 10/27/2022]
Abstract
Additional beneficial effects of angiotensin II type 1 (AT(1)) receptor blockers beyond AT(1) receptor blockade have been highlighted. Irbesartan is reported to act as an antagonist of the monocyte chemoattractant protein-1 (MCP-1) receptor, C-C chemokine receptor 2 (CCR2). We examined the possible synergistic effects of the combination of irbesartan with rosuvastatin on preventing vascular remodeling focusing on the MCP-1/CCR2 pathway. We observed that administration of irbesartan and CCR2 antagonist, propagermanium, at noneffective doses, decreased the neointima with a decrease in PCNA labeling index in the injured mouse femoral artery induced by cuff placement. We also observed that administration of a noneffective dose of rosuvastatin with propagermanium decreased the neointima area, suggesting that the inhibitory effect of rosuvastatin on neointima formation is at least partly attributable to blockade of the MCP-1/CCR2 pathway. Moreover, we demonstrated that the combination of irbesartan with rosuvastatin decreased neointima formation. MCP-1 mRNA level was significantly increased in injured femoral arteries, and administration of irbesartan with rosuvastatin decreased the mRNA levels of MCP-1, TNFα, and IL-1β, and increased PPARγ mRNA expression. These results suggest that the synergistic inhibitory effects of irbesartan with rosuvastatin on neointima formation may involve attenuation of MCP-1/CCR2 signaling.
Collapse
Affiliation(s)
- Kousei Ohshima
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
125
|
Zhao D, McCully BH, Brooks VL. Rosiglitazone improves insulin sensitivity and baroreflex gain in rats with diet-induced obesity. J Pharmacol Exp Ther 2012; 343:206-13. [PMID: 22815534 PMCID: PMC3464031 DOI: 10.1124/jpet.112.194738] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 07/17/2012] [Indexed: 12/24/2022] Open
Abstract
Obesity decreases baroreflex gain (BRG); however, the mechanisms are unknown. We tested the hypothesis that impaired BRG is related to the concurrent insulin resistance, and, therefore, BRG would be improved after treatment with the insulin-sensitizing drug rosiglitazone. Male rats fed a high-fat diet diverged into obesity-prone (OP) and obesity-resistant (OR) groups after 2 weeks. Then, OP and OR rats, as well as control (CON) rats fed a standard diet, were treated daily for 2 to 3 weeks with rosiglitazone (3 or 6 mg/kg) or its vehicle by gavage. Compared with OR and CON rats, conscious OP rats exhibited reductions in BRG (OP, 2.9 ± 0.1 bpm/mm Hg; OR, 4.0 ± 0.2 bpm/mm Hg; CON, 3.9 ± 0.2 bpm/mm Hg; P < 0.05) and insulin sensitivity (hyperinsulinemic euglycemic clamp; OP, 6.8 ± 0.9 mg/kg · min; OR, 22.2 ± 1.2 mg/kg · min; CON, 17.7 ± 0.8 mg/kg · min; P < 0.05), which were well correlated (r(2) = 0.49; P < 0.01). In OP rats, rosiglitazone dose-dependently improved (P < 0.05) insulin sensitivity (12.8 ± 0.6 mg/kg · min at 3 mg/kg; 16.0 ± 1.5 mg/kg · min at 6 mg/kg) and BRG (3.8 ± 0.4 bpm/mm Hg at 3 mg/kg; 5.3 ± 0.7 bpm/mm Hg at 6 mg/kg). However, 6 mg/kg rosiglitazone also increased BRG in OR rats without increasing insulin sensitivity, disrupted the correlation between BRG and insulin sensitivity (r(2) = 0.08), and, in OP and OR rats, elevated BRG relative to insulin sensitivity (analysis of covariance; P < 0.05). Moreover, in OP rats, stimulation of the aortic depressor nerve, to activate central baroreflex pathways, elicited markedly reduced decreases in heart rate and arterial pressure, but these responses were not improved by rosiglitazone. In conclusion, diet-induced obesity impairs BRG via a central mechanism that is related to the concurrent insulin resistance. Rosiglitazone normalizes BRG, but not by improving brain baroreflex processing or insulin sensitivity.
Collapse
Affiliation(s)
- Ding Zhao
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR 97239, USA
| | | | | |
Collapse
|
126
|
Tiyerili V, Müller CF, Fung S, Panek D, Nickenig G, Becher UM. Estrogen improves vascular function via peroxisome-proliferator-activated-receptor-γ. J Mol Cell Cardiol 2012; 53:268-76. [DOI: 10.1016/j.yjmcc.2012.05.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2012] [Revised: 05/04/2012] [Accepted: 05/12/2012] [Indexed: 12/24/2022]
|
127
|
El-Bassossy HM, Abo-Warda SM, Fahmy A. Rosiglitazone, a peroxisome proliferator-activated receptor γ stimulant, abrogates diabetes-evoked hypertension by rectifying abnormalities in vascular reactivity. Clin Exp Pharmacol Physiol 2012; 39:643-9. [DOI: 10.1111/j.1440-1681.2012.05724.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
| | - Shaymaa M Abo-Warda
- Department of Pharmacology; Faculty of Pharmacy; Zagazig University; Zagazig; Egypt
| | - Ahmed Fahmy
- Department of Pharmacology; Faculty of Pharmacy; Zagazig University; Zagazig; Egypt
| |
Collapse
|
128
|
Role of Peroxisome Proliferator-Activated Receptor-γ in Vascular Inflammation. Int J Vasc Med 2012; 2012:508416. [PMID: 22888436 PMCID: PMC3409528 DOI: 10.1155/2012/508416] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2012] [Accepted: 06/08/2012] [Indexed: 12/13/2022] Open
Abstract
Vascular inflammation plays a crucial role in atherosclerosis, and its regulation is important to prevent cerebrovascular and coronary artery disease. The inflammatory process in atherogenesis involves a variety of immune cells including monocytes/macrophages, lymphocytes, dendritic cells, and neutrophils, which all express peroxisome proliferator-activated receptor-γ (PPAR-γ). PPAR-γ is a nuclear receptor and transcription factor in the steroid superfamily and is known to be a key regulator of adipocyte differentiation. Increasing evidence from mainly experimental studies has demonstrated that PPAR-γ activation by endogenous and synthetic ligands is involved in lipid metabolism and anti-inflammatory activity. In addition, recent clinical studies have shown a beneficial effect of thiazolidinediones, synthetic PPAR-γ ligands, on cardiovascular disease beyond glycemic control. These results suggest that PPAR-γ activation is an important regulator in vascular inflammation and is expected to be a therapeutic target in the treatment of atherosclerotic complications. This paper reviews the recent findings of PPAR-γ involvement in vascular inflammation and the therapeutic potential of regulating the immune system in atherosclerosis.
Collapse
|
129
|
Taguchi K, Okada A, Yasui T, Kobayashi T, Ando R, Tozawa K, Kohri K. Pioglitazone, a peroxisome proliferator activated receptor γ agonist, decreases renal crystal deposition, oxidative stress and inflammation in hyperoxaluric rats. J Urol 2012; 188:1002-11. [PMID: 22819112 DOI: 10.1016/j.juro.2012.04.103] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Indexed: 11/16/2022]
Abstract
PURPOSE Kidney stone disease has characteristics similar to those of metabolic syndrome, including inflammation and oxidative stress. The peroxisome proliferator activated receptor γ agonist pioglitazone (AK Scientific, Union, California) is used to treat type 2 diabetes mellitus with an adjunctive effect that improves glycemic control and has anti-inflammatory and antioxidative effects. We investigated the preventive effects of pioglitazone for stone formation in a hyperoxaluric rat model. MATERIALS AND METHODS We divided Sprague-Dawley® rats into a control group, a 1% ethylene glycol group and a 1% ethylene glycol plus 10 mg/kg pioglitazone group. Blood and 24-hour urine samples, and kidney sections were collected on days 7, 14 and 28. We examined crystal formation using Pizzolato staining and polarized light optical microscopy. We also evaluated cell injury, apoptosis and oxidative stress with N-acetyl-β-glucosaminidase, 8-hydroxydeoxyguanosine and TUNEL assay. Expression of crystal and inflammation related genes was examined by immunohistochemistry and quantitative reverse transcriptase-polymerase chain reaction. RESULTS Kidney crystal formation was significantly less in the ethylene glycol plus pioglitazone group than in the ethylene glycol group. Cell injury, apoptosis and oxidative stress markedly decreased after pioglitazone administration. Expression of osteopontin and ED1 for proinflammatory macrophages was lower in the ethylene glycol plus pioglitazone group than in the ethylene glycol group while that of ED2 for anti-inflammatory macrophages was the same in the 2 groups. Linear regression analysis showed a significant change in the correlation coefficient with pioglitazone treatment between Spp1 and Sod1 expression, and the amount of crystals. CONCLUSIONS Pioglitazone suppressed kidney crystal formation through renal tubular cell protection, and antioxidative and anti-inflammatory effects in hyperoxaluric rats.
Collapse
Affiliation(s)
- Kazumi Taguchi
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | | | | | | | | | | | | |
Collapse
|
130
|
Effects of rosiglitazone on the cardiovascular profile in postmenopausal women without diabetes mellitus. Menopause 2012; 19:812-9. [DOI: 10.1097/gme.0b013e3182400ec0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
131
|
Huang JV, Greyson CR, Schwartz GG. PPAR-γ as a therapeutic target in cardiovascular disease: evidence and uncertainty. J Lipid Res 2012; 53:1738-54. [PMID: 22685322 DOI: 10.1194/jlr.r024505] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Peroxisome proliferator-activated receptor γ (PPAR-γ) is a key regulator of fatty acid metabolism, promoting its storage in adipose tissue and reducing circulating concentrations of free fatty acids. Activation of PPAR-γ has favorable effects on measures of adipocyte function, insulin sensitivity, lipoprotein metabolism, and vascular structure and function. Despite these effects, clinical trials of thiazolidinedione PPAR-γ activators have not provided conclusive evidence that they reduce cardiovascular morbidity and mortality. The apparent disparity between effects on laboratory measurements and clinical outcomes may be related to limitations of clinical trials, adverse effects of PPAR-γ activation, or off-target effects of thiazolidinedione agents. This review addresses these issues from a clinician's perspective and highlights several ongoing clinical trials that may help to clarify the therapeutic role of PPAR-γ activators in cardiovascular disease.
Collapse
Affiliation(s)
- Janice V Huang
- Cardiology Section, Denver VA Medical Center, US Department of Veterans Affairs, Denver, CO, USA
| | | | | |
Collapse
|
132
|
Pioglitazone inhibits platelet function and potentiates the effects of aspirin: A prospective observation study. Thromb Res 2012; 129:760-4. [DOI: 10.1016/j.thromres.2011.12.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 12/06/2011] [Accepted: 12/11/2011] [Indexed: 01/13/2023]
|
133
|
Activation of nuclear factor erythroid 2-related factor 2 and PPARγ plays a role in the genistein-mediated attenuation of oxidative stress-induced endothelial cell injury. Br J Nutr 2012; 109:223-35. [PMID: 22716961 DOI: 10.1017/s0007114512001110] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We investigate the cytoprotective effects and the molecular mechanism of genistein in oxidative stress-induced injury using an endothelial cell line (EA.hy926). An oxidative stress model was established by incubating endothelial cells with H₂O₂. According to the present results, genistein pretreatment protected endothelial cells against H₂O₂-induced decreases in cell viability and increases in apoptosis. Genistein also prevented the inhibition of B-cell lymphoma 2 and the activation of caspase-3 induced by H₂O₂. Genistein increased superoxide dismutase (SOD), catalase (CAT) and glutathione (GSH) levels and attenuated the decrease in these antioxidants during oxidative stress. We also found that genistein induced the promoter activity of both nuclear factor erythroid 2-related factor 2 (Nrf2) and PPARγ. Additionally, genistein induced the nuclear translocation of Nrf2 and PPARγ. While genistein caused the up-regulation of both Nrf2 and PPARγ, it also activated and up-regulated the protein expression and transcription of a downstream protein, haem oxygenase-1 (HO-1). Moreover, the use of Nrf2 small interfering RNA transfection and HO-1- or PPARγ-specific antagonists (Znpp and GW9662, respectively) blocked the protective effects of genistein on endothelial cell viability during oxidative stress. Therefore, we conclude that oxidative stress-induced endothelial cell injury can be attenuated by treatment with genistein, which functions via the regulation of the Nrf2 and PPARγ signalling pathway. Additionally, the endogenous antioxidants SOD, CAT and GSH appear to play a role in the antioxidant activity of genistein. The present findings suggest that the beneficial effects of genistein involving the activation of cytoprotective antioxidant genes may represent a novel strategy in the prevention and treatment of cardiovascular endothelial damage.
Collapse
|
134
|
Corthals AP. Multiple sclerosis is not a disease of the immune system. QUARTERLY REVIEW OF BIOLOGY 2012; 86:287-321. [PMID: 22384749 DOI: 10.1086/662453] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Multiple sclerosis is a complex neurodegenerative disease, thought to arise through autoimmunity against antigens of the central nervous system. The autoimmunity hypothesis fails to explain why genetic and environmental risk factors linked to the disease in one population tend to be unimportant in other populations. Despite great advances in documenting the cell and molecular mechanisms underlying MS pathophysiology, the autoimmunity framework has also been unable to develop a comprehensive explanation of the etiology of the disease. I propose a new framework for understanding MS as a dysfunction of the metabolism of lipids. Specifically, the homeostasis of lipid metabolism collapses during acute-phase inflammatory response triggered by a pathogen, trauma, or stress, starting a feedback loop of increased oxidative stress, inflammatory response, and proliferation of cytoxic foam cells that cross the blood brain barrier and both catabolize myelin and prevent remyelination. Understanding MS as a chronic metabolic disorder illuminates four aspects of disease onset and progression: 1) its pathophysiology; 2) genetic susceptibility; 3) environmental and pathogen triggers; and 4) the skewed sex ratio of patients. It also suggests new avenues for treatment.
Collapse
Affiliation(s)
- Angelique P Corthals
- Department of Sciences, John Jay College of Criminal Justice, City University of New York New York, New York 10019, USA.
| |
Collapse
|
135
|
Yu L, Jin X, Yang Y, Cui N, Jiang C. Rosiglitazone inhibits vascular KATP channels and coronary vasodilation produced by isoprenaline. Br J Pharmacol 2012; 164:2064-72. [PMID: 21671900 DOI: 10.1111/j.1476-5381.2011.01539.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE Rosiglitazone is an anti-diabetic drug improving insulin sensitivity and glucose uptake in skeletal muscle and adipose tissues. However, several recent clinical trials suggest that rosiglitazone can increase the risk of cardiovascular ischaemia, although other studies failed to show such risks. Therefore, the effects of rosiglitazone on the coronary circulation and any potential vascular targets need to be elucidated. Here, we show that the vascular isoform of the ATP-sensitive K(+) (K(ATP) ) channel is inhibited by rosiglitazone, impairing physiological regulation of the coronary circulation. EXPERIMENTAL APPROACH The K(IR) 6.1/SUR2B channel was expressed in HEK293 cells and studied in whole-cell and inside-out patch configurations. The Langendorff heart preparation was used to evaluate rosiglitazone in the coronary circulation of wild-type (WT) and K(IR) 6.1-null (Kcnj8(-/-) ) mice. KEY RESULTS K(IR) 6.1/SUR2B channels in HEK cells were inhibited by rosiglitazone in a membrane-delimited manner. This effect was markedly enhanced by sub-micromolar concentrations of glibenclamide and the IC(50) for rosiglitazone fell to 2µM, a therapeutically achievable concentration. In the Langendorff heart preparation rosiglitazone inhibited, concentration-dependently, the coronary vasodilation induced by isoprenaline, without affecting basal coronary tone. Effects of rosiglitazone on coronary perfusion were attenuated by more than 50% in the Kcnj8(-/-) mice, supporting the involvement of K(ATP) channels in this effect of rosiglitazone on the coronary circulation. CONCLUSIONS AND IMPLICATIONS These results indicate that the vascular K(ATP) channel is one of the targets of rosiglitazone action, through which this drug may compromise coronary responses to circulating vasodilators and perhaps also to metabolic stress.
Collapse
Affiliation(s)
- Lei Yu
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA
| | | | | | | | | |
Collapse
|
136
|
Ciborowski M, Teul J, Martin-Ventura JL, Egido J, Barbas C. Metabolomics with LC-QTOF-MS permits the prediction of disease stage in aortic abdominal aneurysm based on plasma metabolic fingerprint. PLoS One 2012; 7:e31982. [PMID: 22384120 PMCID: PMC3286447 DOI: 10.1371/journal.pone.0031982] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 01/16/2012] [Indexed: 11/23/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a permanent and localized aortic dilation, defined as aortic diameter ≥3 cm. It is an asymptomatic but potentially fatal condition because progressive enlargement of the abdominal aorta is spontaneously evolving towards rupture. Biomarkers may help to explain pathological processes of AAA expansion, and allow us to find novel therapeutic strategies or to determine the efficiency of current therapies. Metabolomics seems to be a good approach to find biomarkers of AAA. In this study, plasma samples of patients with large AAA, small AAA, and controls were fingerprinted with LC-QTOF-MS. Statistical analysis was used to compare metabolic fingerprints and select metabolites that showed a significant change. Results presented here reveal that LC-QTOF-MS based fingerprinting of plasma from AAA patients is a very good technique to distinguish small AAA, large AAA, and controls. With the use of validated PLS-DA models it was possible to classify patients according to the disease stage and predict properly the stage of additional AAA patients. Identified metabolites indicate a role for sphingolipids, lysophospholipids, cholesterol metabolites, and acylcarnitines in the development and progression of AAA. Moreover, guanidinosuccinic acid, which mimics nitric oxide in terms of its vasodilatory action, was found as a strong marker of large AAA.
Collapse
Affiliation(s)
- Michal Ciborowski
- CEMBIO (Center for Metabolomics and Bioanalysis), Facultad de Farmacia, Universidad CEU San Pablo, Madrid, Spain
- Department of Physical Chemistry, Medical University of Bialystok, Bialystok, Poland
| | - Joanna Teul
- CEMBIO (Center for Metabolomics and Bioanalysis), Facultad de Farmacia, Universidad CEU San Pablo, Madrid, Spain
- Department of Pharmaceutical Analysis, Medical University of Bialystok, Bialystok, Poland
| | - Jose Luis Martin-Ventura
- Vascular Research Laboratory, IIS-Fundación Jiménez Díaz, Madrid, Spain
- Autónoma University, Madrid, Spain
| | - Jesús Egido
- Vascular Research Laboratory, IIS-Fundación Jiménez Díaz, Madrid, Spain
- Autónoma University, Madrid, Spain
| | - Coral Barbas
- CEMBIO (Center for Metabolomics and Bioanalysis), Facultad de Farmacia, Universidad CEU San Pablo, Madrid, Spain
- * E-mail:
| |
Collapse
|
137
|
Peroxisome proliferator-activated receptor γ agonist troglitazone inhibits high mobility group box 1 expression in endothelial cells via suppressing transcriptional activity of nuclear factor κB and activator protein 1. Shock 2012; 36:228-34. [PMID: 21617575 DOI: 10.1097/shk.0b013e318225b29a] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
High mobility group box 1 (HMGB1), a delayed mediator of proinflammatory cytokines, could initiate and amplify inflammatory responses to infection, injury, and other inflammatory stimuli, and it has emerged as a potential therapeutic target for inflammatory diseases. The overexpression of HMGB1 in endothelial cells has been proved to contribute to the development of these diseases. Because many proinflammatory cytokines expression were suppressed by thiazolidinediones (TZDs), agonists for nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ), whether TZDs can inhibit HMGB1 expression and function is of great interest, however, it remains unknown. Herein, we provide evidence that PPARγ agonist troglitazone, a member of the TZD class, modulates HMGB1 expression in the endothelial cell line EA.hy926 and propose a potential mechanism for that. Results from polymerase chain reaction experiments revealed that PPARγ is expressed in EA.hy926 cells, and it can be activated by troglitazone. Troglitazone inhibited the basal and LPS-stimulated HMGB1 expression at the mRNA level and protein level. A luciferase reporter assay showed that troglitazone inhibited not only the transcriptional activation of the HMGB1 promoter but also activities of heterologous promoters driven by nuclear factor κB (NF-κB) or activator protein 1 (AP-1) response elements. Altogether, these data suggest that NF-κB and AP-1 may participate in the inhibitory effect on HMGB1 transcription induced by troglitazone. Activation of PPARγ by troglitazone is effective for HMGB1 inhibition via suppressing NF-κB and AP-1 transcriptional activity in endothelial cells, which provides a new potential strategy to suppress excessive HMGB1 in inflammatory diseases.
Collapse
|
138
|
Yang FL, Subeq YM, Lee CJ, Lee RP, Peng TC, Harn HJ, Hsu BG. Rosiglitazone protects against severe hemorrhagic shock-induced organ damage in rats. Med Sci Monit 2012; 17:BR282-9. [PMID: 21959602 PMCID: PMC3539481 DOI: 10.12659/msm.881975] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Background Hemorrhagic shock (HS) followed by resuscitation can induce the production of several inflammatory mediators and lead to multiple organ dysfunction. The molecular mechanism of biologic responses to rosiglitazone has an anti-inflammatory effect. The present study was designed to investigate the effects of rosiglitazone on physiopathology and inflammatory mediators after HS in rats. Material/Methods HS was induced in rats by withdrawing 60% of the total blood volume from a femoral artery catheter, immediately followed by intravenous injection of 0.3 mg/kg rosiglitazone. Mean arterial pressure (MAP) and heart rate (HR) were monitored continuously for 12 h. Levels of biochemical parameters, including GOT, GPT, BUN, Cre, LDH, CPK, and lactate were measured at 30 min before induction of HS and 0, 1, 3, 6, 9, and 12 h after HS, while an equal volume of normal saline was replaced as fluid resuscitation. Inflammatory mediators, including tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and monocyte chemoattractant protein-1 (MCP-1), were measured in serum at 1 and 12 h after HS. The kidneys, liver, lungs, and small intestine were removed for histological assessment by hematoxylin and eosin stained at 48 h after HS. Results HS significantly increased blood GOT, GPT, BUN, Cre, LDH, CPK, lactate, glucose, TNF-α, IL-6 and MCP-1 levels, induced tachycardia, and decreased mean arterial pressure (MAP) in rats. Treatment with rosiglitazone improved survival rate, decreased the markers of organ injury, and suppressed the release of TNF-α, IL-6, and MCP-1 after HS in rats. Conclusions Treatment with rosiglitazone suppresses the release of serum TNF-α, IL-6 and MCP-1, and ameliorates HS-induced organ damage in rats.
Collapse
Affiliation(s)
- Fwu-Lin Yang
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | | | | | | | | | | | | |
Collapse
|
139
|
Ashoff A, Qadri F, Eggers R, Jöhren O, Raasch W, Dendorfer A. Pioglitazone Prevents Capillary Rarefaction in Streptozotocin-Diabetic Rats Independently of Glucose Control and Vascular Endothelial Growth Factor Expression. J Vasc Res 2012; 49:260-6. [DOI: 10.1159/000335214] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Accepted: 11/11/2011] [Indexed: 01/13/2023] Open
|
140
|
Sharma B, Singh N. Behavioral and biochemical investigations to explore pharmacological potential of PPAR-gamma agonists in vascular dementia of diabetic rats. Pharmacol Biochem Behav 2011; 100:320-9. [DOI: 10.1016/j.pbb.2011.08.020] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Revised: 07/28/2011] [Accepted: 08/22/2011] [Indexed: 01/04/2023]
|
141
|
Perampaladas K, Gori T, Parker JD. Rosiglitazone Causes Endothelial Dysfunction in Humans. J Cardiovasc Pharmacol Ther 2011; 17:260-5. [DOI: 10.1177/1074248411425490] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Kumar Perampaladas
- Division of Cardiology, Department of Medicine, University of Health Network and Mount Sinai Hospitals, University of Toronto, Toronto, Ontario, Canada
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Tommaso Gori
- Department of Cardiology, University of Mainz, Mainz, Germany
| | - John D. Parker
- Division of Cardiology, Department of Medicine, University of Health Network and Mount Sinai Hospitals, University of Toronto, Toronto, Ontario, Canada
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
142
|
Qu A, Shah YM, Manna SK, Gonzalez FJ. Disruption of endothelial peroxisome proliferator-activated receptor γ accelerates diet-induced atherogenesis in LDL receptor-null mice. Arterioscler Thromb Vasc Biol 2011; 32:65-73. [PMID: 22015658 DOI: 10.1161/atvbaha.111.239137] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Peroxisome proliferator-activated receptor γ (PPARγ) is widely expressed in vessel walls, and it's activation by agonists showed beneficial effects in cardiovascular diseases. However, the role of endothelial cell (EC) PPARγ in atherogenesis is not fully understood. METHODS AND RESULTS To assess the contribution of endothelial-specific PPARγ in atherosclerosis, EC-specific PPARγ disruption and LDL receptor (LDLR) double-knockout (PPARγ(ΔEC)/LDLR(-/-)) mice were developed. When challenged with a high-cholesterol diet for 4 weeks, PPARγ(ΔEC)/LDLR(-/-) mice exhibited severe atherosclerotic lesions compared to either their littermate controls or macrophage-specific PPARγ disruption and LDLR double knockout (PPARγ(ΔMΦ)/LDLR(-/-)) mice. Metabolic analysis showed severe dyslipidemia and significant increase in systolic blood pressure in the PPARγ(ΔEC)/LDLR(-/-) mice. Histological analysis and real-time quantitative PCR suggested an exacerbated inflammation in PPARγ(ΔEC)/LDLR(-/-) mice, as revealed by the increases of proinflammatory gene expression and macrophage infiltration in vivo and in vitro. Furthermore, in vivo endothelial permeability was also increased by endothelial PPARγ disruption. Bone-marrow transplantation studies, which reconstituted hematopoietic PPARγ, demonstrated that the accelerated atherogenesis was due to endothelial PPARγ deficiency. CONCLUSIONS Endothelial PPARγ plays an important protective role in atherogenesis.
Collapse
Affiliation(s)
- Aijuan Qu
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
| | | | | | | |
Collapse
|
143
|
Shen Y, Yuan Z, Yin A, Liu Y, Xiao Y, Wu Y, Wang L, Liang X, Zhao Y, Tian Y, Liu W, Chen T, Kishimoto C. Antiatherogenic effect of pioglitazone on uremic apolipoprotein E knockout mice by modulation of the balance of regulatory and effector T cells. Atherosclerosis 2011; 218:330-8. [DOI: 10.1016/j.atherosclerosis.2011.07.112] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Revised: 07/09/2011] [Accepted: 07/26/2011] [Indexed: 11/16/2022]
|
144
|
Otani H. Oxidative stress as pathogenesis of cardiovascular risk associated with metabolic syndrome. Antioxid Redox Signal 2011; 15:1911-26. [PMID: 21126197 DOI: 10.1089/ars.2010.3739] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Metabolic syndrome (MetS) is characterized by accumulation of visceral fat associated with the clustering of metabolic and pathophysiological cardiovascular risk factors: impaired glucose tolerance, dyslipidemia, and hypertension. Although the definition of MetS is different among countries, visceral obesity is an indispensable component of MetS. A growing body of evidence suggests that increased oxidative stress to adipocytes is central to the pathogenesis of cardiovascular disease in MetS. Increased oxidative stress to adipocytes causes dysregulated expression of inflammation-related adipocytokines in MetS, which contributes to obesity-associated vasculopathy and cardiovascular risk primarily through endothelial dysfunction. The purpose of present review is to unravel the mechanistic link between oxidative stress and cardiovascular risk in MetS, focusing on insulin resistance, hypertension, and atherosclerosis. Then, therapeutic opportunities translated from the bench to bedside will be provided to develop novel strategies to cardiovascular risk factors in MetS.
Collapse
Affiliation(s)
- Hajime Otani
- Second Department of Internal Medicine, Kansai Medical University, 10-15-Fumizono-cho, Moriguchi City, Japan.
| |
Collapse
|
145
|
Yuan X, Zhang Z, Gong K, Zhao P, Qin J, Liu N. Inhibition of reactive oxygen species/extracellular signal-regulated kinases pathway by pioglitazone attenuates advanced glycation end products-induced proliferation of vascular smooth muscle cells in rats. Biol Pharm Bull 2011; 34:618-23. [PMID: 21532147 DOI: 10.1248/bpb.34.618] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Advanced glycation end products (AGEs) have been shown to induce the proliferation of vascular smooth muscle cells (VSMCs) and contribute to atherogenesis and diabetes. In the present study, we investigated the effects of pioglitazone, a peroxisome proliferator activated receptor gamma (PPARγ) agonist, on AGE-induced rat VSMC growth and the underlying mechanism. In cultured rat VSMCs, AGE treatment induced VSMC proliferation in time- and dose-dependent manner, while down-regulated the expression of PPARγ. Pretreatment of pioglitazone not only prevented the down-regulation of PPARγ, but inhibited VSMC proliferation and prevented S-phase entry of cell via a G0-G1 block in the presence of AGEs. Western blotting analysis showed that AGE treatment potentiated to activate extracelluar signal-regulated kinases (ERK1/2) by the induction of intracellular reactive oxygen species (ROS) production, since ROS scavenger N-acetyl-L-cysteine pretreatment significantly inhibited AGE-induced ERK1/2 activation. Further, pretreatment with either N-acetyl-L-cysteine or the inhibitor of ERK1/2 activation suppressed AGE-induced proliferation of VSMCs, suggesting a role of ROS/ERK1/2 signaling. Notably, we demonstrated that pretreatment of pioglitazone significantly attenuated AGE-induced ROS and ERK1/2 activation. Collectively, these results suggest that pioglitazone inhibits AGE-induced VSMC proliferation via increasing PPARγ expression and inhibiting ROS/ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Xiaochen Yuan
- Department of Cardiology, Institute of Cardiovascular Disease of Southeast University, The First People's Hospital of Yangzhou, China
| | | | | | | | | | | |
Collapse
|
146
|
Kang BY, Kleinhenz JM, Murphy TC, Hart CM. The PPARγ ligand rosiglitazone attenuates hypoxia-induced endothelin signaling in vitro and in vivo. Am J Physiol Lung Cell Mol Physiol 2011; 301:L881-91. [PMID: 21926265 DOI: 10.1152/ajplung.00195.2011] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Peroxisome proliferator-activated receptor (PPAR) γ activation attenuates hypoxia-induced pulmonary hypertension (PH) in mice. The current study examined the hypothesis that PPARγ attenuates hypoxia-induced endothelin-1 (ET-1) signaling to mediate these therapeutic effects. To test this hypothesis, human pulmonary artery endothelial cells (HPAECs) were exposed to normoxia or hypoxia (1% O(2)) for 72 h and treated with or without the PPARγ ligand rosiglitazone (RSG, 10 μM) during the final 24 h of exposure. HPAEC proliferation was measured with MTT assays or cell counting, and mRNA and protein levels of ET-1 signaling components were determined. To explore the role of hypoxia-activated transcription factors, selected HPAECs were treated with inhibitors of hypoxia-inducible factor (HIF)-1α (chetomin) or nuclear factor (NF)-κB (caffeic acid phenethyl ester, CAPE). In parallel studies, male C57BL/6 mice were exposed to normoxia (21% O(2)) or hypoxia (10% O(2)) for 3 wk with or without gavage with RSG (10 mg·kg(-1)·day(-1)) for the final 10 days of exposure. Hypoxia increased ET-1, endothelin-converting enzyme-1, and endothelin receptor A and B levels in mouse lung and in HPAECs and increased HPAEC proliferation. Treatment with RSG attenuated hypoxia-induced activation of HIF-1α, NF-κB activation, and ET-1 signaling pathway components. Similarly, treatment with chetomin or CAPE prevented hypoxia-induced increases in HPAEC ET-1 mRNA and protein levels. These findings indicate that PPARγ activation attenuates a program of hypoxia-induced ET-1 signaling by inhibiting activation of hypoxia-responsive transcription factors. Targeting PPARγ represents a novel therapeutic strategy to inhibit enhanced ET-1 signaling in PH pathogenesis.
Collapse
Affiliation(s)
- Bum-Yong Kang
- Department of Medicine, Atlanta Veterans Affairs Medical Centers, GA 30033, USA
| | | | | | | |
Collapse
|
147
|
Abstract
Calcific aortic valve stenosis (CAVS) is a major health problem facing aging societies. The identification of osteoblast-like and osteoclast-like cells in human tissue has led to a major paradigm shift in the field. CAVS was thought to be a passive, degenerative process, whereas now the progression of calcification in CAVS is considered to be actively regulated. Mechanistic studies examining the contributions of true ectopic osteogenesis, nonosseous calcification, and ectopic osteoblast-like cells (that appear to function differently from skeletal osteoblasts) to valvular dysfunction have been facilitated by the development of mouse models of CAVS. Recent studies also suggest that valvular fibrosis, as well as calcification, may play an important role in restricting cusp movement, and CAVS may be more appropriately viewed as a fibrocalcific disease. High-resolution echocardiography and magnetic resonance imaging have emerged as useful tools for testing the efficacy of pharmacological and genetic interventions in vivo. Key studies in humans and animals are reviewed that have shaped current paradigms in the field of CAVS, and suggest promising future areas for research.
Collapse
Affiliation(s)
- Jordan D Miller
- Department of Surgery, Mayo Clinic, Rochester, Minnesota 55905, USA.
| | | | | |
Collapse
|
148
|
Morrison A, Li J. PPAR-γ and AMPK – Advantageous targets for myocardial ischemia/reperfusion therapy. Biochem Pharmacol 2011; 82:195-200. [DOI: 10.1016/j.bcp.2011.04.004] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 04/11/2011] [Accepted: 04/14/2011] [Indexed: 12/25/2022]
|
149
|
NO-Donating NSAIDs, PPARdelta, and Cancer: Does PPARdelta Contribute to Colon Carcinogenesis? PPAR Res 2011; 2008:919572. [PMID: 18528523 PMCID: PMC2408682 DOI: 10.1155/2008/919572] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2008] [Revised: 04/25/2008] [Accepted: 05/06/2008] [Indexed: 12/30/2022] Open
Abstract
The chemopreventive NO-donating NSAIDs (NO-NSAIDs; NSAIDs with an NO-releasing moiety) modulate PPARδ and offer the opportunity to revisit the controversial role of PPARδ in carcinogenesis (several papers report that PPARδ either promotes or inhibits cancer). This review summarizes the pharmacology of NO-NSAIDs, PPARδ cancer biology, and the relationship between the two. In particular, a study of the chemopreventive effect of two isomers of NO-aspirin on intestinal neoplasia in Min mice showed that, compared to wild-type controls, PPARδ is overexpressed in the intestinal mucosa of Min mice; PPARδ responds to
m- and p-NO-ASA proportionally to their antitumor effect (p- > m-). This effect is accompanied by the induction of epithelial cell death, which correlates with the antineoplastic effect of NO-aspirin; and NO-aspirin's effect on PPARδ is specific (no changes in PPARα or PPARγ). Although these data support the notion that PPARδ promotes intestinal carcinogenesis and its inhibition could be therapeutically useful, more work is needed before a firm conclusion is reached.
Collapse
|
150
|
Sain H, Sharma B, Jaggi AS, Singh N. Pharmacological investigations on potential of peroxisome proliferator-activated receptor-gamma agonists in hyperhomocysteinemia-induced vascular dementia in rats. Neuroscience 2011; 192:322-33. [PMID: 21777659 DOI: 10.1016/j.neuroscience.2011.07.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Revised: 06/17/2011] [Accepted: 07/04/2011] [Indexed: 12/25/2022]
Abstract
The present study has been designed to investigate the potential of peroxisome proliferator-activated receptor-gamma ([PPAR]-γ) agonists, pioglitazone, and rosiglitazone in hyperhomocysteinemia-induced vascular dementia of rats. l-methionine was administered for 8 weeks to induce hyperhomocysteinemia and associated vascular dementia. Pioglitazone and rosiglitazone were administered to l-methionine-treated rats for 4 weeks (starting from 5th to 8th weeks of methionine treatment). Donepezil served as a positive control in this study. On 52nd day onward, the animals were exposed to Morris water maze (MWM) for testing learning and memory abilities. Vascular endothelial function, serum nitrite/nitrate levels, brain thiobarbituric acid reactive species (TBARS), brain reduced glutathione (GSH) levels, and brain acetylcholinesterase (AChE) activity were also measured. l-methionine-treated animals have shown impairment of learning, memory, endothelial function, decrease in serum nitrite/nitrate levels, and brain GSH levels along with increase in brain TBARS levels and AChE activity. Pioglitazone, rosiglitazone, and donepezil significantly improved hyperhomocysteinemia-induced impairment of learning, memory, endothelial dysfunction, and changes in various biochemical parameters. It is concluded that pioglitazone and rosiglitazone may be considered as potential pharmacological agents for the management of hyperhomocysteinemia-induced vascular dementia.
Collapse
Affiliation(s)
- H Sain
- Pharmacology Division, Department of Pharmaceutical Sciences and Drug Research, Faculty of Medicine, Punjabi University, Patiala-147002, Punjab, India
| | | | | | | |
Collapse
|