51
|
Tanaka A, Kawaguchi A, Oyama JI, Ishizu T, Ito H, Fukui J, Kondo T, Kuroki S, Nanasato M, Higashi Y, Kaku K, Inoue T, Murohara T, Node K. Differential effect of concomitant antidiabetic agents on carotid atherosclerosis: a subgroup analysis of the PROLOGUE study. Heart Vessels 2018; 34:375-384. [PMID: 30284018 DOI: 10.1007/s00380-018-1275-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Accepted: 09/28/2018] [Indexed: 11/28/2022]
Abstract
Accumulated evidence shows that some antidiabetic agents attenuate the progression of carotid atherosclerosis assessed as intima-media thickness (IMT). Although some studies have demonstrated an inhibitory effect of dipeptidyl peptidase-4 inhibitors on carotid IMT progression, in the PROLOGUE study sitagliptin failed to slow progression relative to conventional therapy for 24 months. We hypothesized that differences in the concomitant antidiabetic agents between the groups have influenced the progression of carotid IMT. We performed a post hoc analysis of the PROLOGUE study using subgroups stratified by concomitant antidiabetic agents. Although no subgroup with any combination of agents in the overall patients showed a significant difference between sitagliptin group and conventional therapy group in the changes from baseline in mean common carotid artery (CCA)-IMT at 24 months, a significant attenuation of mean CCA-IMT progression was observed in the sitagliptin group relative to conventional therapy group only in three combination subgroups aged < 70 years, namely no thiazolidinedione; no thiazolidinedione or biguanide; and no thiazolidinedione, biguanide or α-glucosidase inhibitor, even after adjustment for multiple confounding factors. In the three subgroups, no significant difference between sitagliptin group and conventional therapy group in the changes from baseline in HbA1c at 24 months was detected. Our data suggest that some concomitant agents, whose prescription frequencies were increased in the conventional therapy group, may have masked the inhibitory effect of sitagliptin on carotid IMT progression in the PROLOGUE study.
Collapse
Affiliation(s)
- Atsushi Tanaka
- Department of Cardiovascular Medicine, Saga University, Saga, Japan.
| | | | - Jun-Ichi Oyama
- Department of Cardiovascular Medicine, Saga University, Saga, Japan
| | - Tomoko Ishizu
- Department of Clinical Laboratory Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hiroshi Ito
- Department of Cardiovascular and Respiratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Jun Fukui
- Division of Cardiology, Hokusho Central Hospital, Sasebo, Japan
| | - Taizo Kondo
- Department of Cardiovascular Medicine, Gifu Prefectural Tajimi Hospital, Tajimi, Japan
| | | | - Mamoru Nanasato
- Cardiovascular Center, Japanese Red Cross Nagoya Daini Hospital, Nagoya, Japan
| | - Yukihito Higashi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Kohei Kaku
- Department of General Internal Medicine, Kawasaki Medical School, Kurashiki, Japan
| | - Teruo Inoue
- Department of Cardiovascular Medicine, Dokkyo Medical University, Mibu, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Koichi Node
- Department of Cardiovascular Medicine, Saga University, Saga, Japan.
| |
Collapse
|
52
|
Wang H, Yang Y, Sun X, Tian F, Guo S, Wang W, Tian Z, Jin H, Zhang Z, Tian Y. Sonodynamic therapy-induced foam cells apoptosis activates the phagocytic PPARγ-LXRα-ABCA1/ABCG1 pathway and promotes cholesterol efflux in advanced plaque. Am J Cancer Res 2018; 8:4969-4984. [PMID: 30429880 PMCID: PMC6217053 DOI: 10.7150/thno.26193] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 09/10/2018] [Indexed: 01/09/2023] Open
Abstract
In advanced atherosclerotic plaques, defective efferocytosis of apoptotic foam cells and decreased cholesterol efflux contribute to lesion progression. In our previous study, we demonstrated that 5-aminolevulinic acid (ALA)-mediated sonodynamic therapy (SDT) could induce foam cells apoptosis via the mitochondrial-caspase pathway. In the current research, we sought to explore ALA-SDT-induced apoptosis of phagocytes and the effects of cholesterol efflux and efferocytosis in advanced apoE-/- mice plaque. Methods: apoE-/- mice fed western diet were treated with ALA-SDT and sacrificed at day 1, day 3, day 7 and day 28 post treatment. THP-1 macrophage-derived foam cells were treated with ALA-SDT. 5 hours later, the supernatant was collected and added to fresh foam cells (phagocytes). Then, the lipid area, efferocytosis, cholesterol efflux, anti-inflammatory reactions and PPARγ-LXRα-ABCA1/ABCG1 pathway were detected in plaque in vivo and in phagocytes in vitro. Results: We found that ALA-SDT induced foam cells apoptosis coupled with efferocytosis and upregulation of Mer tyrosine kinase (MerTK) both in vivo and in vitro. The lipid content in plaque decreased as early as 1 day after ALA-SDT and this tendency persisted until 28 days. The enhancement of phagocytes cholesterol efflux was accompanied by an approximately 40% decrease in free cholesterol and a 24% decrease in total cholesterol in vitro. More importantly, anti-inflammatory factors such as TGFβ and IL-10 were upregulated by ALA-SDT treatment. Finally, we found that PPARγ-LXRα-ABCA1/ABCG1 pathway was activated both in vivo and in vitro by ALA-SDT, which could be blocked by PPARγ siRNA. Conclusions: Activation of PPARγ-LXRα-ABCA1/ABCG1 pathway induced by ALA-SDT treatment engages a virtuous cycle that enhances efferocytosis, cholesterol efflux and anti-inflammatory reactions in advanced plaque in vivo and in phagocytes in vitro.
Collapse
|
53
|
An in vitro test system for compounds that modulate human inflammatory macrophage polarization. Eur J Pharmacol 2018; 833:328-338. [DOI: 10.1016/j.ejphar.2018.06.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 06/13/2018] [Accepted: 06/15/2018] [Indexed: 12/22/2022]
|
54
|
Cazzaniga A, Locatelli L, Castiglioni S, Maier J. The Contribution of EDF1 to PPARγ Transcriptional Activation in VEGF-Treated Human Endothelial Cells. Int J Mol Sci 2018; 19:ijms19071830. [PMID: 29933613 PMCID: PMC6073190 DOI: 10.3390/ijms19071830] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 06/14/2018] [Indexed: 12/26/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) is important for maintaining healthy endothelium, which is crucial for vascular integrity. In this paper, we show that VEGF stimulates the nuclear translocation of endothelial differentiation-related factor 1 (EDF1), a highly conserved intracellular protein implicated in molecular events that are pivotal to endothelial function. In the nucleus, EDF1 serves as a transcriptional coactivator of peroxisome proliferator-activated receptor gamma (PPARγ), which has a protective role in the vasculature. Indeed, silencing EDF1 prevents VEGF induction of PPARγ activity as detected by gene reporter assay. Accordingly, silencing EDF1 markedly inhibits the stimulatory effect of VEGF on the expression of FABP4, a PPARγ-inducible gene. As nitric oxide is a marker of endothelial function, it is noteworthy that we report a link between EDF1 silencing, decreased levels of FABP4, and nitric oxide production. We conclude that EDF1 is required for VEGF-induced activation of the transcriptional activity of PPARγ.
Collapse
Affiliation(s)
- Alessandra Cazzaniga
- Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università degli Studi di Milano, I-20157 Milan, Italy.
| | - Laura Locatelli
- Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università degli Studi di Milano, I-20157 Milan, Italy.
| | - Sara Castiglioni
- Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università degli Studi di Milano, I-20157 Milan, Italy.
| | - Jeanette Maier
- Dipartimento di Scienze Biomediche e Cliniche L. Sacco, Università degli Studi di Milano, I-20157 Milan, Italy.
| |
Collapse
|
55
|
Feng L, Liu W, Yang J, Wang Q, Wen S. Effect of Hexadecyl Azelaoyl Phosphatidylcholine on Cardiomyocyte Apoptosis in Myocardial Ischemia-Reperfusion Injury: A Hypothesis. Med Sci Monit 2018; 24:2661-2667. [PMID: 29706617 PMCID: PMC5949054 DOI: 10.12659/msm.907578] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Reperfusion after myocardial ischemia can induce cardiomyocyte death, known as myocardial reperfusion injury. The pathophysiology of the process of reperfusion suggests the confluence multiple pathways. Recent studies have focused on the inflammatory response, which is considered to be the main mechanism during the process of myocardial ischemia-reperfusion injury and can cause cardiomyocyte apoptosis. Peroxisome proliferator-activated receptors gamma activated by endogenous ligands and exogenous ligand can decrease the inflammatory response in cardiomyocytes. Thiazolidinediones are synthetic, high-affinity, selective ligands for peroxisome proliferator-activated receptors gamma, and can inhibit the inflammatory response, decrease myocardial infarct size, and protect cardiac function. However, thiazolidinediones, including rosiglitazone and pioglitazone, can also contribute to adverse cardiovascular events such as congestive heart failure. Therefore, there are some limitations to the use of thiazolidinediones. Most endogenous ligands were of low affinity until hexadecyl azelaoyl phosphatidylcholine was identified as a high-affinity ligand and agonist for peroxisome proliferator-activated receptors gamma. Hexadecyl azelaoyl phosphatidylcholine binds recombinant peroxisome proliferator-activated receptors with an affinity (Kd(app) ≈40 nM) which is equivalent to rosiglitazone. Therefore, hexadecyl azelaoyl phosphatidylcholine is a specific peroxisome proliferator-activated receptors gamma agonist. Given these findings, we hypothesized that the use of hexadecyl azelaoyl phosphatidylcholine can activate the peroxisome proliferator-activated receptors gamma signal pathways and prevent the inflammatory response process of myocardial ischemia-reperfusion injury, with reduced cardiomyocyte apoptosis and death.
Collapse
Affiliation(s)
- Limin Feng
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China (mainland)
| | - Wennan Liu
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin, China (mainland)
| | - Jianzhou Yang
- Department of Preventive Medicine, Changzhi Medical College, Changzhi, Shanxi, China (mainland)
| | - Qing Wang
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin, China (mainland)
| | - Shiwu Wen
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China (mainland)
| |
Collapse
|
56
|
Effects of microRNA-292-5p on myocardial ischemia-reperfusion injury through the peroxisome proliferator-activated receptor-α/-γ signaling pathway. Gene Ther 2018; 25:234-248. [PMID: 29670247 DOI: 10.1038/s41434-018-0014-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 02/10/2018] [Accepted: 02/26/2018] [Indexed: 12/30/2022]
Abstract
Ischemia-reperfusion injury (IRI) is a major cause of cardiac damage following various pathological processes, such as free radical damage and cell apoptosis. This study aims to investigate whether microRNA-292-5p (miR-292-5p) protects against myocardial ischemia-reperfusion injury (IRI) via the peroxisome proliferator-activated receptor (PPAR)-α/-γ signaling pathway in myocardial IRI mice models. Mouse models of myocardial IRI were established. Adult male C57BL/6 mice were divided into different groups. The hemodynamic indexes, levels of related inflammatory factors and serum myocardial enzymes, and malondialdehyde (MDA) content and the activity of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) were detected. The 2,3,5-triphenyltetrazolium chloride (TTC) staining was applied to determine infarct size. TUNEL staining was used to detect cardiomyocyte apoptosis. RT-qPCR and western blotting were performed to measure the related gene expressions. Compared with the model group and the T0070907 + miR-292-5p inhibitor, the miR-292-5p inhibitor group exhibited decreased incidence and duration time of ventricular tachycardia and ventricular fibrillation, serum myocardial enzymes, TNF-α, IL-6, IL-1β, MDA, cardiomyocyte apoptosis, expressions of Bax and p53 in addition to increased SOD and GSH-Px activity, and increased expressions of Bcl-2, PPARα, PPARγ, PLIN5, AQP7, and PCK1. The T0070907 group exhibited opposite results compared to the miR-292-5p inhibitor group. The results indicate that miR-292-5p downregulation protects against myocardial IRI through activation of the PPAR-α/PPAR-γ signaling pathway.
Collapse
|
57
|
Linares I, Farrokhi K, Echeverri J, Kaths JM, Kollmann D, Hamar M, Urbanellis P, Ganesh S, Adeyi OA, Yip P, Selzner M, Selzner N. PPAR-gamma activation is associated with reduced liver ischemia-reperfusion injury and altered tissue-resident macrophages polarization in a mouse model. PLoS One 2018; 13:e0195212. [PMID: 29617419 PMCID: PMC5884549 DOI: 10.1371/journal.pone.0195212] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 03/19/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND PPAR-gamma (γ) is highly expressed in macrophages and its activation affects their polarization. The effect of PPAR-γ activation on Kupffer cells (KCs) and liver ischemia-reperfusion injury (IRI) has not yet been evaluated. We investigated the effect of PPAR-γ activation on KC-polarization and IRI. MATERIALS AND METHODS Seventy percent (70%) liver ischemia was induced for 60mins. PPAR-γ-agonist or vehicle was administrated before reperfusion. PPAR-γ-antagonist was used to block PPAR-γ activation. Liver injury, necrosis, and apoptosis were assessed post-reperfusion. Flow-cytometry determined KC-phenotypes (pro-inflammatory Nitric Oxide +, anti-inflammatory CD206+ and anti-inflammatory IL-10+). RESULTS Liver injury assessed by serum AST was significantly decreased in PPAR-γ-agonist versus control group at all time points post reperfusion (1hr: 3092±105 vs 4469±551; p = 0.042; 6hr: 7041±1160 vs 12193±1143; p = 0.015; 12hr: 5746±328 vs 8608±1259; p = 0.049). Furthermore, liver apoptosis measured by TUNEL-staining was significantly reduced in PPAR-γ-agonist versus control group post reperfusion (1hr:2.46±0.49 vs 6.90±0.85%;p = 0.001; 6hr:26.40±2.93 vs 50.13±8.29%; p = 0.048). H&E staining demonstrated less necrosis in PPAR-γ-agonist versus control group (24hr:26.66±4.78 vs 45.62±4.57%; p = 0.032). The percentage of pro-inflammatory NO+ KCs was significantly lower at all post reperfusion time points in the PPAR-γ-agonist versus control group (1hr:28.49±4.99 vs 53.54±9.15%; p = 0.040; 6hr:5.51±0.54 vs 31.12±9.58%; p = 0.009; 24hr:4.15±1.50 vs 17.10±4.77%; p = 0.043). In contrast, percentage of anti-inflammatory CD206+ KCs was significantly higher in PPAR-γ-agonist versus control group prior to IRI (8.62±0.96 vs 4.88 ±0.50%; p = 0.04). Administration of PPAR-γ-antagonist reversed the beneficial effects on AST, apoptosis, and pro-inflammatory NO+ KCs. CONCLUSION PPAR-γ activation reduces IRI and decreases the pro-inflammatory NO+ Kupffer cells. PPAR-γ activation can become an important tool to improve outcomes in liver surgery through decreasing the pro-inflammatory phenotype of KCs and IRI.
Collapse
Affiliation(s)
- Ivan Linares
- Multi Organ Transplant Program, Toronto General Hospital, Toronto, ON, Canada
- Consejo Nacional de Ciencia y Tecnología, México City, México
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Kaveh Farrokhi
- Multi Organ Transplant Program, Toronto General Hospital, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Juan Echeverri
- Multi Organ Transplant Program, Toronto General Hospital, Toronto, ON, Canada
| | - Johan Moritz Kaths
- Multi Organ Transplant Program, Toronto General Hospital, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Dagmar Kollmann
- Multi Organ Transplant Program, Toronto General Hospital, Toronto, ON, Canada
| | - Matyas Hamar
- Multi Organ Transplant Program, Toronto General Hospital, Toronto, ON, Canada
| | - Peter Urbanellis
- Multi Organ Transplant Program, Toronto General Hospital, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Sujani Ganesh
- Multi Organ Transplant Program, Toronto General Hospital, Toronto, ON, Canada
| | - Oyedele A. Adeyi
- Department of Pathology, Toronto General Hospital, Toronto, ON, Canada
| | - Paul Yip
- Laboratory of Medicine and Pathobiology, Toronto General Hospital, Toronto, ON, Canada
| | - Markus Selzner
- Multi Organ Transplant Program, Toronto General Hospital, Toronto, ON, Canada
| | - Nazia Selzner
- Multi Organ Transplant Program, Toronto General Hospital, Toronto, ON, Canada
- * E-mail:
| |
Collapse
|
58
|
Ateyya H, Nader MA, El-Sherbeeny NA. Beneficial effects of rosiglitazone and losartan combination in diabetic rats. Can J Physiol Pharmacol 2018; 96:215-220. [PMID: 28892640 DOI: 10.1139/cjpp-2017-0332] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Diabetes with vascular complication needs strict interventions to retard possible serious complications. This research estimated the possible interaction of rosiglitazone (RGN) with losartan (Los) in diabetic rats. Male Sprague-Dawley rats were randomly divided into nondiabetic rats, diabetic rats, and diabetic rats that received RGN, Los, or a combination of RGN and Los. Measurement of serum glucose, vascular adhesion molecule-1, interleukin-6, tumor necrosis factor-α, aortic lipid peroxide (malondialdehyde), glutathione, superoxide dismutase, and total nitrate/nitrite levels was done. Also, the effects of RGN on the relaxation created by acetylcholine and sodium nitroprusside, contraction of isolated aortic rings provoked by phenylephrine and angiotensin II were determined. Results revealed that RGN or Los had a vasodilating effect to variable degrees indicated by enhanced effects on both acetylcholine-induced relaxation and the antagonistic effect on angiotensin II and phenylephrine-stimulated contraction of diabetic aortas with significant amelioration in serum glucose, vascular adhesion molecule-1, interleukin-6, and tumor necrosis factor-α levels and aortic oxidant/antioxidant balance. Treatment of diabetic rats with a combination of RGN and Los produced a more pronounced effect on the measured parameters compared to the diabetic, RGN-, and Los-treated groups. These findings point out the beneficial effects of RGN and Los combination in diabetic rats.
Collapse
Affiliation(s)
- Hayam Ateyya
- a College of Pharmacy, Taibah University, El-Madinah El-Munawarah, Saudi Arabia
- b Department of Clinical Pharmacology, Faculty of Medicine, Cairo University, Egypt
| | - Manar A Nader
- a College of Pharmacy, Taibah University, El-Madinah El-Munawarah, Saudi Arabia
- c Faculty of Pharmacy, Mansoura University, Egypt
| | - Nagla A El-Sherbeeny
- a College of Pharmacy, Taibah University, El-Madinah El-Munawarah, Saudi Arabia
- d Department of Clinical Pharmacology, Faculty of Medicine, Suez Canal University, Egypt
| |
Collapse
|
59
|
Altered Circadian Timing System-Mediated Non-Dipping Pattern of Blood Pressure and Associated Cardiovascular Disorders in Metabolic and Kidney Diseases. Int J Mol Sci 2018; 19:ijms19020400. [PMID: 29385702 PMCID: PMC5855622 DOI: 10.3390/ijms19020400] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 01/12/2018] [Accepted: 01/20/2018] [Indexed: 12/15/2022] Open
Abstract
The morning surge in blood pressure (BP) coincides with increased cardiovascular (CV) events. This strongly suggests that an altered circadian rhythm of BP plays a crucial role in the development of CV disease (CVD). A disrupted circadian rhythm of BP, such as the non-dipping type of hypertension (i.e., absence of nocturnal BP decline), is frequently observed in metabolic disorders and chronic kidney disease (CKD). The circadian timing system, controlled by the central clock in the suprachiasmatic nucleus of the hypothalamus and/or by peripheral clocks in the heart, vasculature, and kidneys, modulates the 24 h oscillation of BP. However, little information is available regarding the molecular and cellular mechanisms of an altered circadian timing system-mediated disrupted dipping pattern of BP in metabolic disorders and CKD that can lead to the development of CV events. A more thorough understanding of this pathogenesis could provide novel therapeutic strategies for the management of CVD. This short review will address our and others' recent findings on the molecular mechanisms that may affect the dipping pattern of BP in metabolic dysfunction and kidney disease and its association with CV disorders.
Collapse
|
60
|
Choi JY, Ryu J, Kim HJ, Song JW, Jeon JH, Lee DH, Oh DJ, Gweon DG, Oh WY, Yoo H, Park K, Kim JW. Therapeutic Effects of Targeted PPARɣ Activation on Inflamed High-Risk Plaques Assessed by Serial Optical Imaging In Vivo. Am J Cancer Res 2018; 8:45-60. [PMID: 29290792 PMCID: PMC5743459 DOI: 10.7150/thno.20885] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 09/27/2017] [Indexed: 12/16/2022] Open
Abstract
Rationale: Atherosclerotic plaque is a chronic inflammatory disorder involving lipid accumulation within arterial walls. In particular, macrophages mediate plaque progression and rupture. While PPARγ agonist is known to have favorable pleiotropic effects on atherogenesis, its clinical application has been very limited due to undesirable systemic effects. We hypothesized that the specific delivery of a PPARγ agonist to inflamed plaques could reduce plaque burden and inflammation without systemic adverse effects. Methods: Herein, we newly developed a macrophage mannose receptor (MMR)-targeted biocompatible nanocarrier loaded with lobeglitazone (MMR-Lobe), which is able to specifically activate PPARγ pathways within inflamed high-risk plaques, and investigated its anti-atherogenic and anti-inflammatory effects both in in vitro and in vivo experiments. Results: MMR-Lobe had a high affinity to macrophage foam cells, and it could efficiently promote cholesterol efflux via LXRα-, ABCA1, and ABCG1 dependent pathways, and inhibit plaque protease expression. Using in vivo serial optical imaging of carotid artery, MMR-Lobe markedly reduced both plaque burden and inflammation in atherogenic mice without undesirable systemic effects. Comprehensive analysis of en face aorta by ex vivo imaging and immunostaining well corroborated the in vivo findings. Conclusion: MMR-Lobe was able to activate PPARγ pathways within high-risk plaques and effectively reduce both plaque burden and inflammation. This novel targetable PPARγ activation in macrophages could be a promising therapeutic strategy for high-risk plaques.
Collapse
|
61
|
Qian X, Guo D, Zhou H, Qiu J, Wang J, Shen C, Guo Z, Xu Y, Dong C. Interactions Between PPARG and AGTR1 Gene Polymorphisms on the Risk of Hypertension in Chinese Han Population. Genet Test Mol Biomarkers 2017; 22:90-97. [PMID: 29266977 DOI: 10.1089/gtmb.2017.0141] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS To explore the interactions between PPARG and AGTR1 polymorphisms and their associations with hypertension in the Chinese Han population. METHODS Seven single nucleotide polymorphisms (SNPs) of the PPARG gene and five SNPs of the AGTR1 gene were selected and genotyped in 1591 unrelated Chinese Han adults. The SNPAssoc package of R was used to analyze the associations between the selected SNPs and hypertension. The potential gene-gene interactions between PPARG and AGTR1 genes were tested by model-based multifactor dimensionality reduction (MB-MDR). RESULTS The frequencies of the C allele of rs3856806 and the G allele of rs13433696 in the PPARG gene were significantly lower in hypertensive subjects, whereas the A allele of rs9817428 in the PPARG gene was much higher in hypertensives. In addition, individuals with T allele of rs2933249 in the AGTR1 gene displayed a significantly decreased risk of hypertension. MB-MDR analyses suggested that the two-locus model (rs9817428 and rs2933249) and the three-locus model (rs9817428, rs3856806, and rs2933249) were significantly associated with a decreased risk of hypertension. Moreover, among the eight SNPs not individually associated with hypertension (rs12631819, rs2920502, rs1175543, and rs2972164 in the PPARG gene, and rs2638360, rs1492100, rs5182, and rs275646 in the AGTR1 gene), the two-locus model involving rs12631819 and rs5182 demonstrated increased susceptibility to hypertension, and the five-locus model involving rs12631819, rs2920502, rs2972164, rs5182, and rs2638360 demonstrated a significantly decreased risk of hypertension. CONCLUSION Polymorphisms in both the PPARG and AGTR1 genes were found to be significantly associated with hypertension. Moreover, there were significant gene-gene interactions identified between the PPARG and AGTR1 genes in relation to hypertension susceptibility in the Chinese Han population.
Collapse
Affiliation(s)
- Xiaoyan Qian
- 1 Department of Epidemiology and Statistics, School of Public Health, Jiangsu Key Laboratory and Translational Medicine for Geriatric Disease, Medical College of Soochow University , Suzhou, China
| | - Daoxia Guo
- 1 Department of Epidemiology and Statistics, School of Public Health, Jiangsu Key Laboratory and Translational Medicine for Geriatric Disease, Medical College of Soochow University , Suzhou, China
| | - Hui Zhou
- 2 Suzhou Industrial Park Centers for Disease Control and Prevention , Suzhou, China
| | - Jing Qiu
- 1 Department of Epidemiology and Statistics, School of Public Health, Jiangsu Key Laboratory and Translational Medicine for Geriatric Disease, Medical College of Soochow University , Suzhou, China
| | - Jie Wang
- 1 Department of Epidemiology and Statistics, School of Public Health, Jiangsu Key Laboratory and Translational Medicine for Geriatric Disease, Medical College of Soochow University , Suzhou, China
| | - Chong Shen
- 3 Department of Epidemiology and Statistics, School of Public Health, Nanjing Medical University , Nanjing, China
| | - Zhirong Guo
- 1 Department of Epidemiology and Statistics, School of Public Health, Jiangsu Key Laboratory and Translational Medicine for Geriatric Disease, Medical College of Soochow University , Suzhou, China
| | - Yong Xu
- 1 Department of Epidemiology and Statistics, School of Public Health, Jiangsu Key Laboratory and Translational Medicine for Geriatric Disease, Medical College of Soochow University , Suzhou, China
| | - Chen Dong
- 1 Department of Epidemiology and Statistics, School of Public Health, Jiangsu Key Laboratory and Translational Medicine for Geriatric Disease, Medical College of Soochow University , Suzhou, China
| |
Collapse
|
62
|
Cai W, Yang T, Liu H, Han L, Zhang K, Hu X, Zhang X, Yin KJ, Gao Y, Bennett MVL, Leak RK, Chen J. Peroxisome proliferator-activated receptor γ (PPARγ): A master gatekeeper in CNS injury and repair. Prog Neurobiol 2017; 163-164:27-58. [PMID: 29032144 DOI: 10.1016/j.pneurobio.2017.10.002] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 10/06/2017] [Accepted: 10/08/2017] [Indexed: 01/06/2023]
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is a widely expressed ligand-modulated transcription factor that governs the expression of genes involved in inflammation, redox equilibrium, trophic factor production, insulin sensitivity, and the metabolism of lipids and glucose. Synthetic PPARγ agonists (e.g. thiazolidinediones) are used to treat Type II diabetes and have the potential to limit the risk of developing brain injuries such as stroke by mitigating the influence of comorbidities. If brain injury develops, PPARγ serves as a master gatekeeper of cytoprotective stress responses, improving the chances of cellular survival and recovery of homeostatic equilibrium. In the acute injury phase, PPARγ directly restricts tissue damage by inhibiting the NFκB pathway to mitigate inflammation and stimulating the Nrf2/ARE axis to neutralize oxidative stress. During the chronic phase of acute brain injuries, PPARγ activation in injured cells culminates in the repair of gray and white matter, preservation of the blood-brain barrier, reconstruction of the neurovascular unit, resolution of inflammation, and long-term functional recovery. Thus, PPARγ lies at the apex of cell fate decisions and exerts profound effects on the chronic progression of acute injury conditions. Here, we review the therapeutic potential of PPARγ in stroke and brain trauma and highlight the novel role of PPARγ in long-term tissue repair. We describe its structure and function and identify the genes that it targets. PPARγ regulation of inflammation, metabolism, cell fate (proliferation/differentiation/maturation/survival), and many other processes also has relevance to other neurological diseases. Therefore, PPARγ is an attractive target for therapies against a number of progressive neurological disorders.
Collapse
Affiliation(s)
- Wei Cai
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tuo Yang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Huan Liu
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Lijuan Han
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Kai Zhang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Xiaoming Hu
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai 200032, China; Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh PA, USA
| | - Xuejing Zhang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Ke-Jie Yin
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Michael V L Bennett
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA.
| | - Jun Chen
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai 200032, China; Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh PA, USA.
| |
Collapse
|
63
|
Matoba T, Koga JI, Nakano K, Egashira K, Tsutsui H. Nanoparticle-mediated drug delivery system for atherosclerotic cardiovascular disease. J Cardiol 2017; 70:206-211. [DOI: 10.1016/j.jjcc.2017.03.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 03/02/2017] [Indexed: 01/26/2023]
|
64
|
Sanders WG, Li H, Zhuplatov I, He Y, Kim SE, Cheung AK, Agarwal J, Terry CM. Autologous fat transplants to deliver glitazone and adiponectin for vasculoprotection. J Control Release 2017; 264:237-246. [PMID: 28867378 DOI: 10.1016/j.jconrel.2017.08.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 08/06/2017] [Accepted: 08/29/2017] [Indexed: 10/18/2022]
Abstract
The insulin sensitizing glitazone drugs, rosiglitazone (ROS) and pioglitazone (PGZ) both have anti-proliferative and anti-inflammatory effects and induce adipose tissue (fat) to produce the vaso-protective protein adiponectin. Stenosis due to intimal hyperplasia development often occurs after placement of arteriovenous synthetic grafts used for hemodialysis. This work was performed to characterize the in vitro and in vivo effects of ROS or PGZ incorporation in fat and to determine if fat/PGZ depots could decrease vascular hyperplasia development in a porcine model of hemodialysis arteriovenous graft stenosis. Powdered ROS or PGZ (6-6000μM) was mixed with fat explants and cultured. Drug release from fat was quantified by HPLC/MS/MS, and adiponectin and monocyte chemotactic protein-1 (MCP-1) levels in culture media were measured by ELISA. The effect of conditioned media from the culture of fat with ROS or PGZ on i) platelet-derived growth factor-BB (PDGF-BB)-stimulated proliferation of human venous smooth muscle cells (SMC) was measured by a DNA-binding assay, and ii) lipopolysaccharide (LPS)-induced human monocyte release of tumor necrosis factor-alpha (TNFα) was assessed by ELISA. In a porcine model, pharmacokinetics of PGZ from fat depots transplanted perivascular to jugular vein were assessed by HPLC/MS/MS, and retention of the fat depot was monitored by MRI. A porcine model of synthetic graft placed between carotid artery and ipsilateral jugular vein was used to assess effects of PGZ/fat depots on vascular hyperplasia development. Both ROS and PGZ significantly induced the release of adiponectin and inhibited release of MCP-1 from the fat. TNF production from monocytes stimulated with LPS was inhibited 50-70% in the presence of media conditioned by fat alone or fat and either drug. The proliferation of SMC was inhibited in the presence of media conditioned by fat/ROS cultures. Fat explants placed perivascular to the external jugular vein were retained, as confirmed by MRI at one week after placement. PGZ was detected in the fat depot, in the external jugular vein wall and in adjacent tissue at clinically relevant levels, whereas levels in plasma were below detection. External jugular vein exposed to fat incorporated with PGZ had increased adiponectin expression compared to vein exposed to fat alone. However, the development of hyperplasia within the arteriovenous synthetic grafts was unchanged by treatment with fat/PGZ depots compared to no treatment.
Collapse
Affiliation(s)
- William G Sanders
- Division of Nephrology & Hypertension, Department of Internal Medicine, University of Utah, 295 Chipeta Way, Salt Lake City, UT, USA
| | - Huan Li
- Division of Nephrology & Hypertension, Department of Internal Medicine, University of Utah, 295 Chipeta Way, Salt Lake City, UT, USA
| | - Ilya Zhuplatov
- Division of Nephrology & Hypertension, Department of Internal Medicine, University of Utah, 295 Chipeta Way, Salt Lake City, UT, USA
| | - Yuxia He
- Division of Nephrology & Hypertension, Department of Internal Medicine, University of Utah, 295 Chipeta Way, Salt Lake City, UT, USA
| | - Seong-Eun Kim
- Department of Radiology and Imaging Science, Utah Center for Advanced Imaging Research, University of Utah, 729 Arapeen Dr., Salt Lake City, UT, USA
| | - Alfred K Cheung
- Division of Nephrology & Hypertension, Department of Internal Medicine, University of Utah, 295 Chipeta Way, Salt Lake City, UT, USA; Medical Service, Veterans Affairs Salt Lake City Healthcare System, 500 Foothill Dr., 151N, Salt Lake City, UT, USA
| | - Jayant Agarwal
- Division of Plastic and Reconstructive Surgery, School of Medicine, University of Utah, 30 N. 1900 E. 3B400, Salt Lake City, UT, USA
| | - Christi M Terry
- Division of Nephrology & Hypertension, Department of Internal Medicine, University of Utah, 295 Chipeta Way, Salt Lake City, UT, USA.
| |
Collapse
|
65
|
Ku YH, Cho BJ, Kim MJ, Lim S, Park YJ, Jang HC, Choi SH. Rosiglitazone increases endothelial cell migration and vascular permeability through Akt phosphorylation. BMC Pharmacol Toxicol 2017; 18:62. [PMID: 28854981 PMCID: PMC5577739 DOI: 10.1186/s40360-017-0169-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Accepted: 08/11/2017] [Indexed: 02/06/2023] Open
Abstract
Background Thiazolidinediones (TZDs), peroxisome proliferator-activated receptor-γ (PPAR-γ) agonists, exhibit anti-inflammatory and antioxidant properties and inhibit endothelial inflammation and dysfunction, which is anti-atherogenic. However, fluid retention, which may lead to congestive heart failure and peripheral edema, is also a concern, which may result from endothelial cell leakage. In the current study, we examined the effects of PPAR-γ agonists on vascular endothelial cell migration and permeability in order to determine its underlying mechanisms. Methods We used rosiglitazone and conducted cell migration assay and permeability assay using HUVEC cells and measured vascular permeability and leakage in male C57BL/6 mice. Results Rosiglitazone significantly promoted endothelial cell migration and induced permeability via activation of phosphatidylinositol-3-kinase (PI3K) – Akt or protein kinase C (PKC)β. In addition, rosiglitazone increased vascular endothelial growth factor (VEGF) expression and suppressed expression of tight junction proteins (JAM-A and ZO-1), which might promote neovascularization and vascular leakage. These phenomena were reduced by Akt inhibition. Conclusions Vascular endothelial cell migration and permeability change through Akt phosphorylation might be a mechanism of induced fluid retention and peripheral tissue edema by TZD.
Collapse
Affiliation(s)
- Yun Hyi Ku
- Department of Internal Medicine, Korea Cancer Center Hospital, Seoul, South Korea
| | - Bong-Jun Cho
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Min Joo Kim
- Department of Internal Medicine, Korea Cancer Center Hospital, Seoul, South Korea
| | - Soo Lim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Young Joo Park
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Hak C Jang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Sung Hee Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea. .,, 166 Gumi-dong, Bundang-gu, Seongnam-si, Gyeonggi-do, 463-707, Republic of Korea.
| |
Collapse
|
66
|
Bonnet F, Scheen AJ. Impact of glucose-lowering therapies on risk of stroke in type 2 diabetes. DIABETES & METABOLISM 2017; 43:299-313. [PMID: 28522196 DOI: 10.1016/j.diabet.2017.04.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 04/21/2017] [Indexed: 02/07/2023]
Abstract
Patients with type 2 diabetes (T2D) have an increased risk of stroke compared with people without diabetes. However, the effects of glucose-lowering drugs on risk of ischaemic stroke in T2D have been less extensively investigated than in coronary heart disease. Some evidence, including the UKPDS, has suggested a reduced risk of stroke with metformin, although the number of studies is limited. Inhibition of the KATP channels increases ischaemic brain lesions in animals. This is in agreement with a recent meta-analysis showing an increased risk of stroke with sulphonylureas vs. various comparators as both mono- and combination therapy. Pioglitazone can prevent recurrence of stroke in patients with previous stroke, as already shown in PROactive, although results are less clear for first strokes. As for DPP-4 inhibitors, there was a non-significant trend towards benefit for stroke, whereas a possible increased risk of stroke with SGLT2 inhibitors-and in particular, empagliflozin in the EMPA-REG OUTCOME trial-has been suggested and requires clarification. Experimental results support a potential protective effect of GLP-1 receptor agonists against stroke that has, at least in part, been translated to clinical benefits in T2D patients in the LEADER and SUSTAIN-6 trials. Further interventional studies are now warranted to confirm the effects of glucose-lowering agents on risk of stroke in patients with T2D. In summary, the effects of antidiabetic drugs on risk of stroke appear to be heterogeneous, with some therapies (pioglitazone, GLP-1 receptor agonists) conferring possible protection against ischaemic stroke, other classes showing a neutral impact (DPP-4 inhibitors, insulin) and some glucose-lowering agents being associated with an increased risk of stroke (sulphonylureas, possibly SGLT2 inhibitors, high-dose insulin in the presence of insulin resistance).
Collapse
Affiliation(s)
- F Bonnet
- Centre Hospitalier Universitaire de Rennes, Université Rennes 1, Rennes, France; INSERM U1018, Villejuif, France.
| | - A J Scheen
- Division of Clinical Pharmacology, Centre for Interdisciplinary Research on Medicines (CIRM), University of Liège, Liège, Belgium; Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine, CHU, Liège, Belgium
| |
Collapse
|
67
|
Fan W, Waizenegger W, Lin CS, Sorrentino V, He MX, Wall CE, Li H, Liddle C, Yu RT, Atkins AR, Auwerx J, Downes M, Evans RM. PPARδ Promotes Running Endurance by Preserving Glucose. Cell Metab 2017; 25:1186-1193.e4. [PMID: 28467934 PMCID: PMC5492977 DOI: 10.1016/j.cmet.2017.04.006] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/27/2017] [Accepted: 04/09/2017] [Indexed: 10/19/2022]
Abstract
Management of energy stores is critical during endurance exercise; a shift in substrate utilization from glucose toward fat is a hallmark of trained muscle. Here we show that this key metabolic adaptation is both dependent on muscle PPARδ and stimulated by PPARδ ligand. Furthermore, we find that muscle PPARδ expression positively correlates with endurance performance in BXD mouse reference populations. In addition to stimulating fatty acid metabolism in sedentary mice, PPARδ activation potently suppresses glucose catabolism and does so without affecting either muscle fiber type or mitochondrial content. By preserving systemic glucose levels, PPARδ acts to delay the onset of hypoglycemia and extends running time by ∼100 min in treated mice. Collectively, these results identify a bifurcated PPARδ program that underlies glucose sparing and highlight the potential of PPARδ-targeted exercise mimetics in the treatment of metabolic disease, dystrophies, and, unavoidably, the enhancement of athletic performance.
Collapse
Affiliation(s)
- Weiwei Fan
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Wanda Waizenegger
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Chun Shi Lin
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Vincenzo Sorrentino
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Ming-Xiao He
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Christopher E Wall
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Hao Li
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Christopher Liddle
- Storr Liver Centre, Westmead Institute for Medical Research and Sydney Medical School, University of Sydney, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Ruth T Yu
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Annette R Atkins
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Johan Auwerx
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| | - Ronald M Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Howard Hughes Medical Institute, Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| |
Collapse
|
68
|
Han L, Shen WJ, Bittner S, Kraemer FB, Azhar S. PPARs: regulators of metabolism and as therapeutic targets in cardiovascular disease. Part II: PPAR-β/δ and PPAR-γ. Future Cardiol 2017; 13:279-296. [PMID: 28581362 PMCID: PMC5941699 DOI: 10.2217/fca-2017-0019] [Citation(s) in RCA: 182] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 03/21/2017] [Indexed: 02/06/2023] Open
Abstract
The PPARs are a subfamily of three ligand-inducible transcription factors, which belong to the superfamily of nuclear hormone receptors. In mammals, the PPAR subfamily consists of three members: PPAR-α, PPAR-β/δ and PPAR-γ. PPARs control the expression of a large number of genes involved in metabolic homeostasis, lipid, glucose and energy metabolism, adipogenesis and inflammation. PPARs regulate a large number of metabolic pathways that are implicated in the pathogenesis of metabolic diseases such as metabolic syndrome, Type 2 diabetes mellitus, nonalcoholic fatty liver disease and cardiovascular disease. The aim of this review is to provide up-to-date information about the biochemical and metabolic actions of PPAR-β/δ and PPAR-γ, the therapeutic potential of their agonists currently under clinical development and the cardiovascular disease outcome of clinical trials of PPAR-γ agonists, pioglitazone and rosiglitazone.
Collapse
Affiliation(s)
- Lu Han
- Geriatrics Research, Education & Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
- Division of Endocrinology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Wen-Jun Shen
- Geriatrics Research, Education & Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
- Division of Endocrinology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Stefanie Bittner
- Geriatrics Research, Education & Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Fredric B Kraemer
- Geriatrics Research, Education & Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
- Division of Endocrinology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Salman Azhar
- Geriatrics Research, Education & Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
- Division of Endocrinology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
69
|
Chaudhry A, Carthan KA, Kang BY, Calvert J, Sutliff RL, Michael Hart C. PPARγ attenuates hypoxia-induced hypertrophic transcriptional pathways in the heart. Pulm Circ 2017; 7:98-107. [PMID: 28680569 PMCID: PMC5448534 DOI: 10.1086/689749] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 09/15/2016] [Indexed: 02/01/2023] Open
Abstract
Chronic hypoxia-induced pulmonary hypertension (PH) is characterized by increased pressure and resistance in the pulmonary vasculature and hypertrophy of the right ventricle (RV). The transcription factors, nuclear factor activated T-cells (NFAT), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB/p65) contribute to RV hypertrophy (RVH). Because peroxisome proliferator-activated receptor gamma (PPARγ) activation attenuates hypoxia-induced PH and RVH, we hypothesized that PPARγ inhibits activation of RV hypertrophic transcriptional signaling mechanisms. C57BL/6J mice were exposed to normoxia (21% O2) or hypoxia (10% O2) for 21 days. During the final 10 days of exposure, selected mice were treated with the PPARγ ligand, pioglitazone. RV systolic pressure (RVSP) and RVH were measured, and NFATc2 and NF-kB/p65 protein levels were measured in RV and LV nuclear and cytosolic fractions. Cardiomyocyte hypertrophy was assessed with wheatgerm agglutinin staining. NFAT activation was also examined with luciferase reporter mice and analysis of protein levels of selected transcriptional targets. Chronic-hypoxia increased: (1) RVH, RVSP, and RV cardiomyocyte hypertrophy; (2) NFATc2 and NF-κB activation in RV nuclear homogenates; (3) RV and LV NFAT luciferase activity; and (4) RV protein levels of brain natriuretic peptide (BNP) and β-myosin heavy chain (β-MyHC). Treatment with pioglitazone attenuated hypoxia-induced increases in both RV and LV NFAT luciferase activity. Chronic hypoxia caused sustained RV NFATc2 and NF-κB activation. Pioglitazone attenuated PH, RVH, cardiomyocyte hypertrophy, and activation of RV hypertrophic signaling and also attenuated LV NFAT activation. PPARγ favorably modulates signaling derangements in the heart as well as in the pulmonary vascular wall.
Collapse
Affiliation(s)
- Abubakr Chaudhry
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center and Emory University, Atlanta, GA, USA
| | - Kristal A Carthan
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center and Emory University, Atlanta, GA, USA
| | - Bum-Yong Kang
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center and Emory University, Atlanta, GA, USA
| | - John Calvert
- Department of Surgery, Emory University, Atlanta, GA, USA
| | - Roy L Sutliff
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center and Emory University, Atlanta, GA, USA
| | - C Michael Hart
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Atlanta Veterans Affairs Medical Center and Emory University, Atlanta, GA, USA
| |
Collapse
|
70
|
Perrucci GL, Rurali E, Gowran A, Pini A, Antona C, Chiesa R, Pompilio G, Nigro P. Vascular smooth muscle cells in Marfan syndrome aneurysm: the broken bricks in the aortic wall. Cell Mol Life Sci 2017; 74:267-277. [PMID: 27535662 PMCID: PMC11107581 DOI: 10.1007/s00018-016-2324-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 07/14/2016] [Accepted: 08/02/2016] [Indexed: 01/22/2023]
Abstract
Marfan syndrome (MFS) is a connective tissue disorder with multiple organ manifestations. The genetic cause of this syndrome is the mutation of the FBN1 gene, encoding the extracellular matrix (ECM) protein fibrillin-1. This genetic alteration leads to the degeneration of microfibril structures and ECM integrity in the tunica media of the aorta. Indeed, thoracic aortic aneurysm and dissection represent the leading cause of death in MFS patients. To date, the most effective treatment option for this pathology is the surgical substitution of the damaged aorta. To highlight novel therapeutic targets, we review the molecular mechanisms related to MFS etiology in vascular smooth muscle cells, the foremost cellular type involved in MFS pathogenesis.
Collapse
Affiliation(s)
- Gianluca L Perrucci
- Department of Clinical Sciences and Community Health, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Via C. Parea 4, 20138, Milan, Italy
| | - Erica Rurali
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Via C. Parea 4, 20138, Milan, Italy
| | - Aoife Gowran
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Via C. Parea 4, 20138, Milan, Italy
| | - Alessandro Pini
- Department of Cardiology, Marfan Clinic®, "Luigi Sacco" University of Milan, Via G.B. Grassi 74, 20157, Milan, Italy
| | - Carlo Antona
- Cardiovascular Surgery Department, "Luigi Sacco" University of Milan, Via G.B. Grassi 74, 20157, Milan, Italy
- FoRCardioLab, "Luigi Sacco" University of Milan, Via G.B. Grassi 74, 20157, Milan, Italy
| | - Roberto Chiesa
- Department of Vascular Surgery, San Raffaele Scientific Institute Hospital, Vita-Salute University, Milan, Italy
| | - Giulio Pompilio
- Department of Clinical Sciences and Community Health, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy.
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Via C. Parea 4, 20138, Milan, Italy.
- Department of Cardiovascular Surgery, Centro Cardiologico Monzino-IRCCS, Via C. Parea 4, 20138, Milan, Italy.
| | - Patrizia Nigro
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Via C. Parea 4, 20138, Milan, Italy.
| |
Collapse
|
71
|
Di Mise A, Wang YX, Zheng YM. Role of Transcription Factors in Pulmonary Artery Smooth Muscle Cells: An Important Link to Hypoxic Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 967:13-32. [PMID: 29047078 DOI: 10.1007/978-3-319-63245-2_2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hypoxia, namely a lack of oxygen in the blood, induces pulmonary vasoconstriction and vasoremodeling, which serve as essential pathologic factors leading to pulmonary hypertension (PH). The underlying molecular mechanisms are uncertain; however, pulmonary artery smooth muscle cells (PASMCs) play an essential role in hypoxia-induced pulmonary vasoconstriction, vasoremodeling, and PH. Hypoxia causes oxidative damage to DNAs, proteins, and lipids. This damage (oxidative stress) modulates the activity of ion channels and elevates the intracellular calcium concentration ([Ca2+]i, Ca2+ signaling) of PASMCs. The oxidative stress and increased Ca2+ signaling mutually interact with each other, and synergistically results in a variety of cellular responses. These responses include functional and structural abnormalities of mitochondria, sarcoplasmic reticulum, and nucleus; cell contraction, proliferation, migration, and apoptosis, as well as generation of vasoactive substances, inflammatory molecules, and growth factors that mediate the development of PH. A number of studies reveal that various transcription factors (TFs) play important roles in hypoxia-induced oxidative stress, disrupted PAMSC Ca2+ signaling and the development and progress of PH. It is believed that in the pathogenesis of PH, hypoxia facilitates these roles by mediating the expression of multiple genes. Therefore, the identification of specific genes and their transcription factors implicated in PH is necessary for the complete understanding of the underlying molecular mechanisms. Moreover, this identification may aid in the development of novel and effective therapeutic strategies for PH.
Collapse
Affiliation(s)
- Annarita Di Mise
- Department of Molecular & Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA
| | - Yong-Xiao Wang
- Department of Molecular & Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA.
| | - Yun-Min Zheng
- Department of Molecular & Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA.
| |
Collapse
|
72
|
Goltsman I, Khoury EE, Winaver J, Abassi Z. Does Thiazolidinedione therapy exacerbate fluid retention in congestive heart failure? Pharmacol Ther 2016; 168:75-97. [PMID: 27598860 DOI: 10.1016/j.pharmthera.2016.09.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The ever-growing global burden of congestive heart failure (CHF) and type 2 diabetes mellitus (T2DM) as well as their co-existence necessitate that anti-diabetic pharmacotherapy will modulate the cardiovascular risk inherent to T2DM while complying with the accompanying restrictions imposed by CHF. The thiazolidinedione (TZD) family of peroxisome proliferator-activated receptor γ (PPARγ) agonists initially provided a promising therapeutic option in T2DM owing to anti-diabetic efficacy combined with pleiotropic beneficial cardiovascular effects. However, the utility of TZDs in T2DM has declined in the past decade, largely due to concomitant adverse effects of fluid retention and edema formation attributed to salt-retaining effects of PPARγ activation on the nephron. Presumably, the latter effects are potentially deleterious in the context of pre-existing fluid retention in CHF. However, despite a considerable body of evidence on mechanisms responsible for TZD-induced fluid retention suggesting that this class of drugs is rightfully prohibited from use in CHF patients, there is a paucity of experimental and clinical studies that investigate the effects of TZDs on salt and water homeostasis in the CHF setting. In an attempt to elucidate whether TZDs actually exacerbate the pre-existing fluid retention in CHF, our review summarizes the pathophysiology of fluid retention in CHF. Moreover, we thoroughly review the available data on TZD-induced fluid retention and proposed mechanisms in animals and patients. Finally, we will present recent studies challenging the common notion that TZDs worsen renal salt and water retention in CHF.
Collapse
Affiliation(s)
- Ilia Goltsman
- Department of Physiology, Biophysics and Systems Biology, The Bruce Rappaport, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Emad E Khoury
- Department of Physiology, Biophysics and Systems Biology, The Bruce Rappaport, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Joseph Winaver
- Department of Physiology, Biophysics and Systems Biology, The Bruce Rappaport, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Zaid Abassi
- Department of Physiology, Biophysics and Systems Biology, The Bruce Rappaport, Rappaport Faculty of Medicine, Technion, Haifa, Israel; Department of Laboratory Medicine, Rambam Human Health Care Campus, Haifa, Israel.
| |
Collapse
|
73
|
PPAR Ligands Function as Suppressors That Target Biological Actions of HMGB1. PPAR Res 2016; 2016:2612743. [PMID: 27563308 PMCID: PMC4985574 DOI: 10.1155/2016/2612743] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 06/21/2016] [Accepted: 06/28/2016] [Indexed: 01/12/2023] Open
Abstract
High mobility group box 1 (HMGB1), which has become one of the most intriguing molecules in inflammatory disorders and cancers and with which ligand-activated peroxisome proliferator-activated receptors (PPARs) are highly associated, is considered as a therapeutic target. Of particular interest is the fact that certain PPAR ligands have demonstrated their potent anti-inflammatory activities and potential anticancer effects. In this review article we summarize recent experimental evidence that PPAR ligands function as suppressors that target biological actions of HMGB1, including intracellular expression, receptor signaling cascades, and extracellular secretion of HMGB1 in cell lines and/or animal models. We also propose the possible mechanisms underlying PPAR involvement in inflammatory disorders and discuss the future therapeutic value of PPAR ligands targeting HMGB1 molecule for cancer prevention and treatment.
Collapse
|
74
|
Mahendra J, Parthiban PS, Mahendra L, Balakrishnan A, Shanmugam S, Junaid M, Romanos GE. Evidence Linking the Role of Placental Expressions of Peroxisome Proliferator-Activated Receptor-γ and Nuclear Factor-Kappa B in the Pathogenesis of Preeclampsia Associated With Periodontitis. J Periodontol 2016; 87:962-70. [DOI: 10.1902/jop.2016.150677] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
75
|
Nakano Y, Matoba T, Tokutome M, Funamoto D, Katsuki S, Ikeda G, Nagaoka K, Ishikita A, Nakano K, Koga JI, Sunagawa K, Egashira K. Nanoparticle-Mediated Delivery of Irbesartan Induces Cardioprotection from Myocardial Ischemia-Reperfusion Injury by Antagonizing Monocyte-Mediated Inflammation. Sci Rep 2016; 6:29601. [PMID: 27403534 PMCID: PMC4939605 DOI: 10.1038/srep29601] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/21/2016] [Indexed: 12/19/2022] Open
Abstract
Myocardial ischemia-reperfusion (IR) injury limits the therapeutic effect of early reperfusion therapy for acute myocardial infarction (AMI), in which the recruitment of inflammatory monocytes plays a causative role. Here we develop bioabsorbable poly-lactic/glycolic acid (PLGA) nanoparticles incorporating irbesartan, an angiotensin II type 1 receptor blocker with a peroxisome proliferator-activated receptor (PPAR)γ agonistic effect (irbesartan-NP). In a mouse model of IR injury, intravenous PLGA nanoparticles distribute to the IR myocardium and monocytes in the blood and in the IR heart. Single intravenous treatment at the time of reperfusion with irbesartan-NP (3.0 mg kg(-1) irbesartan), but not with control nanoparticles or irbesartan solution (3.0 mg kg(-1)), inhibits the recruitment of inflammatory monocytes to the IR heart, and reduces the infarct size via PPARγ-dependent anti-inflammatory mechanisms, and ameliorates left ventricular remodeling 21 days after IR. Irbesartan-NP is a novel approach to treat myocardial IR injury in patients with AMI.
Collapse
Affiliation(s)
- Yasuhiro Nakano
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Tetsuya Matoba
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Masaki Tokutome
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Daiki Funamoto
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Shunsuke Katsuki
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Gentaro Ikeda
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kazuhiro Nagaoka
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Ayako Ishikita
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kaku Nakano
- Department of Cardiovascular Research, Development, and Translational Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Jun-Ichiro Koga
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kenji Sunagawa
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kensuke Egashira
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan.,Department of Cardiovascular Research, Development, and Translational Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| |
Collapse
|
76
|
Ansari RA, Husain K, Rizvi SAA. Role of Transcription Factors in Steatohepatitis and Hypertension after Ethanol: The Epicenter of Metabolism. Biomolecules 2016; 6:29. [PMID: 27348013 PMCID: PMC5039415 DOI: 10.3390/biom6030029] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 05/25/2016] [Accepted: 06/08/2016] [Indexed: 02/07/2023] Open
Abstract
Chronic alcohol consumption induces multi-organ damage, including alcoholic liver disease (ALD), pancreatitis and hypertension. Ethanol and ethanol metabolic products play a significant role in the manifestation of its toxicity. Ethanol metabolizes to acetaldehyde and produces reduced nicotinamide adenine dinucleotide (NADH) by cytosolic alcohol dehydrogenase. Ethanol metabolism mediated by cytochrome-P450 2E1 causes oxidative stress due to increased production of reactive oxygen species (ROS). Acetaldehyde, increased redox cellular state and ROS activate transcription factors, which in turn activate genes for lipid biosynthesis and offer protection of hepatocytes from alcohol toxicity. Sterol regulatory element binding proteins (SREBPs) and peroxisome proliferator activated-receptors (PPARs) are two key lipogenic transcription factors implicated in the development of fatty liver in alcoholic and non-alcoholic steatohepatitis. SREBP-1 is activated in the livers of chronic ethanol abusers. An increase in ROS activates nuclear factor erythroid-2-related factor-2 (Nrf2) and hypoxia inducible factor (HIF) to provide protection to hepatocytes from ethanol toxicity. Under ethanol exposure, due to increased gut permeability, there is release of gram-negative bacteria-derived lipopolysaccharide (LPS) from intestine causing activation of immune response. In addition, the metabolic product, acetaldehyde, modifies the proteins in hepatocyte, which become antigens inviting auto-immune response. LPS activates macrophages, especially the liver resident macrophages, Kupffer cells. These Kupffer cells and circulating macrophages secrete various cytokines. The level of tumor necrosis factor-α (TNFα), interleukin-1beta (IL-1β), IL-6, IL-8 and IL-12 have been found elevated among chronic alcoholics. In addition to elevation of these cytokines, the peripheral iron (Fe(2+)) is also mobilized. An increased level of hepatic iron has been observed among alcoholics. Increased ROS, IL-1β, acetaldehyde, and increased hepatic iron, all activate nuclear factor-kappa B (NF-κB) transcription factor. Resolution of increased reactive oxygen species requires increased expression of genes responsible for dismutation of increased ROS which is partially achieved by IL-6 mediated activation of signal transducers and activators of transcription 3 (STAT3). In addition to these transcription factors, activator protein-1 may also be activated in hepatocytes due to its association with resolution of increased ROS. These transcription factors are central to alcohol-mediated hepatotoxicity.
Collapse
Affiliation(s)
- Rais A Ansari
- Department of Pharmaceutical Sciences, College of Pharmacy, Health Professions Division, Nova Southeastern University, 3200 S University Drive, Fort Lauderdale, FL 33328, USA.
| | - Kazim Husain
- Department of Physiology, Pharmacology and Toxicology, Ponce School of Medicine, P.O. Box 7004, Ponce, PR 00732-2575, USA.
| | - Syed A A Rizvi
- Department of Pharmaceutical Sciences, College of Pharmacy, Health Professions Division, Nova Southeastern University, 3200 S University Drive, Fort Lauderdale, FL 33328, USA.
| |
Collapse
|
77
|
Kukida M, Mogi M, Ohshima K, Nakaoka H, Iwanami J, Kanno H, Tsukuda K, Chisaka T, Min LJ, Wang XL, Bai HY, Shan BS, Higaki A, Yamauchi T, Okura T, Higaki J, Horiuchi M. Angiotensin II Type 2 Receptor Inhibits Vascular Intimal Proliferation With Activation of PPARγ. Am J Hypertens 2016; 29:727-36. [PMID: 26471325 DOI: 10.1093/ajh/hpv168] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 09/24/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Angiotensin II type 2 (AT2) receptor stimulation could exert beneficial effects on vascular remodeling. Previously, we reported that AT2 receptor stimulation ameliorated insulin resistance in diabetic mice accompanied by PPARγ activation which also plays a variety of crucial roles in the vasculature. Therefore, this study aimed to investigate the vascular protective effect of the AT2 receptor with activation of PPARγ involving AT2 receptor-interacting protein (ATIP). METHODS AND RESULTS Vascular injury was induced by polyethylene-cuff placement around the femoral artery in C57BL/6J mice. Treatment with compound 21 (C21), an AT2 receptor agonist, decreased neointimal formation, cell proliferation, and the mRNA levels of monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor (TNF)-α, and interleukin-1β, and phosphorylation of nuclear factor-kappa B, and increased PPARγ DNA-binding activity in the injured artery, whereas these inhibitory effects of C21 were attenuated by co-treatment with a PPARγ antagonist, GW9662. Treatment of vascular smooth muscle cells (VSMC) with C21 prepared from smAT2 transgenic mice, which highly express the AT2 receptor in VSMC, increased both PPARγ activity and its DNA-binding activity determined by dual-luciferase assay and electrophoresis mobility shift assay (EMSA), respectively. We observed that ATIP was involved in PPARγ complex formation, and that transfection of siRNA of ATIP1 attenuated the AT2 receptor-mediated increase in PPARγ activity in VSMC. In response to AT2 receptor stimulation, ATIP was translocated from the plasma membrane to the nucleus. CONCLUSIONS Our results suggest a new mechanism by which AT2 receptor stimulation activates PPARγ, thereby resulting in amelioration of vascular intimal proliferation, and that ATIP plays an important role in AT2 receptor-mediated PPARγ activation.
Collapse
Affiliation(s)
- Masayoshi Kukida
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan; Department of Cardiology, Pulmonology, Hypertension and Nephrology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Masaki Mogi
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan;
| | - Kousei Ohshima
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan; Department of Cardiology, Pulmonology, Hypertension and Nephrology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Hirotomo Nakaoka
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Jun Iwanami
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Harumi Kanno
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Kana Tsukuda
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Toshiyuki Chisaka
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan; Department of Pediatrics, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Li-Juan Min
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Xiao-Li Wang
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Hui-Yu Bai
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Bao-Shuai Shan
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Akinori Higaki
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan; Department of Cardiology, Pulmonology, Hypertension and Nephrology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Toshifumi Yamauchi
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan; Department of Pediatrics, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Takafumi Okura
- Department of Cardiology, Pulmonology, Hypertension and Nephrology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Jitsuo Higaki
- Department of Cardiology, Pulmonology, Hypertension and Nephrology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Masatsugu Horiuchi
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| |
Collapse
|
78
|
Xu M, Huang Y, Xie L, Peng K, Ding L, Lin L, Wang P, Hao M, Chen Y, Sun Y, Qi L, Wang W, Ning G, Bi Y. Diabetes and Risk of Arterial Stiffness: A Mendelian Randomization Analysis. Diabetes 2016; 65:1731-40. [PMID: 26953161 DOI: 10.2337/db15-1533] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 02/29/2016] [Indexed: 11/13/2022]
Abstract
We aimed to explore the causal association between type 2 diabetes (T2D) and increased arterial stiffness. We performed a Mendelian randomization (MR) analysis in 11,385 participants from a well-defined community study in Shanghai during 2011-2013. We genotyped 34 T2D-associated common variants identified in East Asians and created a genetic risk score (GRS). We assessed arterial stiffness noninvasively with the measurement of brachial-ankle pulse wave velocity (baPWV). We used the instrumental variable (IV) estimator to qualify the causal relationship between T2D and increased arterial stiffness. We found each 1-SD increase in T2D_GRS was associated with 6% higher risk in increased arterial stiffness (95% CI 1.01, 1.12), after adjustment of other metabolic confounders. Using T2D_GRS as the IV, we demonstrated a causal relationship between T2D and arterial stiffening (odds ratio 1.24, 95% CI 1.06, 1.47; P = 0.008). When categorizing the genetic loci according to their effect on insulin secretion or resistance, we found genetically determined decrease in insulin secretion was associated with increase in baPWV (βIV = 122.3 cm/s, 95% CI 41.9, 204.6; P = 0.0005). In conclusion, our results provide evidence supporting a causal association between T2D and increased arterial stiffness in a Chinese population.
Collapse
Affiliation(s)
- Min Xu
- State Key Laboratory of Medical Genomics, Key Laboratory for Endocrine and Metabolic Diseases of Ministry of Health, National Clinical Research Center for Metabolic Diseases, Collaborative Innovation Center of Systems Biomedicine, and Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ya Huang
- State Key Laboratory of Medical Genomics, Key Laboratory for Endocrine and Metabolic Diseases of Ministry of Health, National Clinical Research Center for Metabolic Diseases, Collaborative Innovation Center of Systems Biomedicine, and Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lan Xie
- Department of Biomedical Engineering, Medical Systems Biology Research Center, Tsinghua University School of Medicine, Beijing, China
| | - Kui Peng
- State Key Laboratory of Medical Genomics, Key Laboratory for Endocrine and Metabolic Diseases of Ministry of Health, National Clinical Research Center for Metabolic Diseases, Collaborative Innovation Center of Systems Biomedicine, and Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Ding
- State Key Laboratory of Medical Genomics, Key Laboratory for Endocrine and Metabolic Diseases of Ministry of Health, National Clinical Research Center for Metabolic Diseases, Collaborative Innovation Center of Systems Biomedicine, and Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Lin
- State Key Laboratory of Medical Genomics, Key Laboratory for Endocrine and Metabolic Diseases of Ministry of Health, National Clinical Research Center for Metabolic Diseases, Collaborative Innovation Center of Systems Biomedicine, and Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Po Wang
- State Key Laboratory of Medical Genomics, Key Laboratory for Endocrine and Metabolic Diseases of Ministry of Health, National Clinical Research Center for Metabolic Diseases, Collaborative Innovation Center of Systems Biomedicine, and Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingli Hao
- State Key Laboratory of Medical Genomics, Key Laboratory for Endocrine and Metabolic Diseases of Ministry of Health, National Clinical Research Center for Metabolic Diseases, Collaborative Innovation Center of Systems Biomedicine, and Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhong Chen
- State Key Laboratory of Medical Genomics, Key Laboratory for Endocrine and Metabolic Diseases of Ministry of Health, National Clinical Research Center for Metabolic Diseases, Collaborative Innovation Center of Systems Biomedicine, and Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yimin Sun
- Department of Biomedical Engineering, Medical Systems Biology Research Center, Tsinghua University School of Medicine, Beijing, China National Engineering Research Center for Beijing Biochip Technology, Beijing, China
| | - Lu Qi
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA
| | - Weiqing Wang
- State Key Laboratory of Medical Genomics, Key Laboratory for Endocrine and Metabolic Diseases of Ministry of Health, National Clinical Research Center for Metabolic Diseases, Collaborative Innovation Center of Systems Biomedicine, and Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- State Key Laboratory of Medical Genomics, Key Laboratory for Endocrine and Metabolic Diseases of Ministry of Health, National Clinical Research Center for Metabolic Diseases, Collaborative Innovation Center of Systems Biomedicine, and Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yufang Bi
- State Key Laboratory of Medical Genomics, Key Laboratory for Endocrine and Metabolic Diseases of Ministry of Health, National Clinical Research Center for Metabolic Diseases, Collaborative Innovation Center of Systems Biomedicine, and Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China Department of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
79
|
Maekawa H, Serrone JC, Tjahjadi M, Hernesniemi J. RETRACTED ARTICLE: The role of estrogen on the pathology of cerebral aneurysms. Expert Rev Neurother 2016; 16:927-35. [DOI: 10.1080/14737175.2016.1189827] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
80
|
Jin X, Liu L, Zhou Z, Ge J, Yao T, Shen C. Pioglitazone alleviates inflammation in diabetic mice fed a high-fat diet via inhibiting advanced glycation end-product-induced classical macrophage activation. FEBS J 2016; 283:2295-308. [PMID: 27062545 DOI: 10.1111/febs.13735] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 02/22/2016] [Accepted: 04/06/2016] [Indexed: 11/30/2022]
Abstract
Classically activated macrophages (M1) are associated with inflammation in diabetic patients. Inflammation is a known risk factor in diabetes. The present study tested the hypothesis that pioglitazone (PIO) alleviates inflammation in diabetic mice fed a high-fat diet by inhibiting advanced glycation end-product (AGE)-induced classical macrophage activation. It was found that AGE treatment promoted the transcription of pro-inflammatory molecules and M1 surface markers, whereas PIO increased the expression of anti-inflammatory genes and decreased the expression of pro-inflammatory mediators in bone marrow-derived macrophages (BMDMs) in a dose-dependent manner. Furthermore, pretreatment with PIO abrogated the effects of AGE on pro-inflammatory markers and partly inhibited AGE-induced nuclear factor-κB (NF-κB) activation. PIO treatment partly reduced the inflammatory phenotype in diabetic ApoE(-/-) mice, and significantly reduced NF-κB activation in plaques. Therefore, we conclude that PIO blocks classical activation of macrophages and attenuates inflammation in mouse models of diabetes.
Collapse
Affiliation(s)
- Xian Jin
- Department of Cardiology, Central Hospital of Minhang District, Shanghai, China.,Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Liang Liu
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Zhong'e Zhou
- Department of Cardiology, Central Hospital of Minhang District, Shanghai, China.,Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Junhua Ge
- Department of Cardiology, The Affiliated Hospital of Qingdao University, China
| | - Tongqing Yao
- Department of Cardiology, Tongling People's Hospital, China
| | - Chengxing Shen
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, China
| |
Collapse
|
81
|
Rolland M, Li X, Sellier Y, Martin H, Perez-Berezo T, Rauwel B, Benchoua A, Bessières B, Aziza J, Cenac N, Luo M, Casper C, Peschanski M, Gonzalez-Dunia D, Leruez-Ville M, Davrinche C, Chavanas S. PPARγ Is Activated during Congenital Cytomegalovirus Infection and Inhibits Neuronogenesis from Human Neural Stem Cells. PLoS Pathog 2016; 12:e1005547. [PMID: 27078877 PMCID: PMC4831785 DOI: 10.1371/journal.ppat.1005547] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 03/12/2016] [Indexed: 11/25/2022] Open
Abstract
Congenital infection by human cytomegalovirus (HCMV) is a leading cause of permanent sequelae of the central nervous system, including sensorineural deafness, cerebral palsies or devastating neurodevelopmental abnormalities (0.1% of all births). To gain insight on the impact of HCMV on neuronal development, we used both neural stem cells from human embryonic stem cells (NSC) and brain sections from infected fetuses and investigated the outcomes of infection on Peroxisome Proliferator-Activated Receptor gamma (PPARγ), a transcription factor critical in the developing brain. We observed that HCMV infection dramatically impaired the rate of neuronogenesis and strongly increased PPARγ levels and activity. Consistent with these findings, levels of 9-hydroxyoctadecadienoic acid (9-HODE), a known PPARγ agonist, were significantly increased in infected NSCs. Likewise, exposure of uninfected NSCs to 9-HODE recapitulated the effect of infection on PPARγ activity. It also increased the rate of cells expressing the IE antigen in HCMV-infected NSCs. Further, we demonstrated that (1) pharmacological activation of ectopically expressed PPARγ was sufficient to induce impaired neuronogenesis of uninfected NSCs, (2) treatment of uninfected NSCs with 9-HODE impaired NSC differentiation and (3) treatment of HCMV-infected NSCs with the PPARγ inhibitor T0070907 restored a normal rate of differentiation. The role of PPARγ in the disease phenotype was strongly supported by the immunodetection of nuclear PPARγ in brain germinative zones of congenitally infected fetuses (N = 20), but not in control samples. Altogether, our findings reveal a key role for PPARγ in neurogenesis and in the pathophysiology of HCMV congenital infection. They also pave the way to the identification of PPARγ gene targets in the infected brain. Congenital infection by human cytomegalovirus (HCMV) might result in permanent neurological sequelae, including sensorineural deafness, cerebral palsies or devastating neurodevelopmental abnormalities. Infants with such sequelae represent about 0.1% of all live births (>5500 per year in the USA). Given the considerable health and societal burden, a better insight on disease pathogenesis is urgently needed to design new therapeutic or prognostic tools. Here, we studied the impact of HCMV on neuronal development, using human neural progenitors (NSC) as a disease model. In particular, we investigated the outcome of infection on Peroxisome Proliferator-Activated Receptor gamma (PPARγ, a key protein in the regulation of metabolism, inflammation and cell differentiation. We disclosed that HCMV infection strongly increases levels and activity of PPARγ in NSCs. In vitro experiments showed that PPARγ activity inhibits the differentiation of NSCs into neurons. We also found increased PPARγ expression in brains of in utero infected fetuses, but not in controls, suggesting that PPARγ is a key effector of HCMV infection also in vivo. Our study provides new insights on the pathogenesis of HCMV infection and paves the way to the discovery of PPARγ-related molecules secreted in the infected brain.
Collapse
Affiliation(s)
- Maude Rolland
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
| | - Xiaojun Li
- Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Yann Sellier
- Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Hélène Martin
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
| | - Teresa Perez-Berezo
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
| | - Benjamin Rauwel
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
| | | | - Bettina Bessières
- Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jacqueline Aziza
- Département d'Anatomie Pathologique, IUCT-Oncopole, Toulouse, France
| | - Nicolas Cenac
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
| | - Minhua Luo
- Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Charlotte Casper
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
- Neonatal Unit, Children’s Hospital, Toulouse, France
| | - Marc Peschanski
- I-STEM, INSERM U861, AFM, Evry, France
- CECS, UEVE U861, Evry, France
| | - Daniel Gonzalez-Dunia
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
| | - Marianne Leruez-Ville
- Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Christian Davrinche
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
| | - Stéphane Chavanas
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
- * E-mail:
| |
Collapse
|
82
|
Tsukahara T, Haniu H, Matsuda Y, Murakmi-Murofushi K. Short-term treatment with a 2-carba analog of cyclic phosphatidic acid induces lowering of plasma cholesterol levels in ApoE-deficient mice. Biochem Biophys Res Commun 2016; 473:107-113. [PMID: 27012212 DOI: 10.1016/j.bbrc.2016.03.060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 03/15/2016] [Indexed: 10/22/2022]
Abstract
Plasma cholesterol levels are associated with an increased risk of developing atherosclerosis. An elevated low-density lipoprotein cholesterol (LDL-C) level is a hallmark of hypercholesterolemia in metabolic syndrome. Our previous study suggested that when acetylated LDL (AC-LDL) was co-applied with a PPARγ agonist, rosiglitazone (ROSI), many oil red O-positive macrophages could be observed. However, addition of cyclic phosphatidic acid (cPA) to ROSI-stimulated macrophages completely abolished oil red O-stained cells, indicating that cPA inhibits PPARγ-regulated AC-LDL uptake. This study aimed to determine whether metabolically stabilized cPA, in the form of a carba-derivative of cPA (2ccPA), could reduce plasma cholesterol levels and affect the expression of genes related to atherosclerosis in apolipoprotein E-knockout (apoE(-/-)) mice. 2ccPA reduced LDL-C levels in these mice (n = 3) from 460 to 330 mg/ml, from 420 to 350 mg/ml, and 420 to 281 mg/ml under a western-type diet. 2ccPA also reduced expression of lipid metabolism-related genes, cytokines, and chemokines in ApoE-deficient mice on a high-fat diet. Taken together, these results suggest that 2ccPA governs anti-atherogenic activities in the carotid arteries of apoE-deficient mice.
Collapse
Affiliation(s)
- Tamotsu Tsukahara
- Department of Molecular Pharmacology and Neuroscience, Nagasaki University Graduate School of Biomedical Sciences, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan.
| | - Hisao Haniu
- Institute for Biomedical Sciences, Shinshu University Interdisciplinary Cluster for Cutting Edge Research, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Yoshikazu Matsuda
- Clinical Pharmacology Educational Center, Nihon Pharmaceutical University, Ina-machi, Saitama 362-0806, Japan
| | - Kimiko Murakmi-Murofushi
- Endowed Research Division of Human Welfare Sciences, Ochanomizu University, 2-1-1, Ohtsuka, Bunkyo-ku, Tokyo 112-8610, Japan
| |
Collapse
|
83
|
Behl T, Kaur I, Kotwani A. Role of endocannabinoids in the progression of diabetic retinopathy. Diabetes Metab Res Rev 2016; 32:251-9. [PMID: 26379208 DOI: 10.1002/dmrr.2710] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 08/04/2015] [Accepted: 09/15/2015] [Indexed: 12/17/2022]
Abstract
In the past decades, the role of numerous factors in the pathophysiology of diabetic retinopathy has been explored, following which marked progress has been made in developing several novel therapeutic options, such as anti-vascular endothelial growth factor, anti-tumor necrosis factor-alpha and various other anti-inflammatory and anti-angiogenic agents, for the treatment of diabetic retinopathy. However, the involvement of endocannabinoid system in its pathogenesis has not been much explored. This review aims at unveiling every aspect of association of the endocannabinoid system and its interactions with various physiological and pathological pathways to induce disease progression. The various alterations induced by endocannabinoids, such as anandamide and 2-arachidonylglycerol, in retina during hyperglycaemia clearly demonstrate and verify their involvement in aggravating the pathological conditions, hence leading to the progression of diabetic retinopathy. Exploring this involvement furthermore, in greater depths, might be beneficial in acknowledging and understanding the hidden aspects of the pathogenesis of this complication even better and might provide a therapeutically beneficial alternative target to combat and restrict its progression amongst diabetic patients.
Collapse
Affiliation(s)
- Tapan Behl
- Department of Pharmacology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Ishneet Kaur
- Department of Pharmacy, Chandigarh College of Pharmacy, Mohali, Punjab, India
| | - Anita Kotwani
- Department of Pharmacology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| |
Collapse
|
84
|
Nakashiro S, Matoba T, Umezu R, Koga JI, Tokutome M, Katsuki S, Nakano K, Sunagawa K, Egashira K. Pioglitazone-Incorporated Nanoparticles Prevent Plaque Destabilization and Rupture by Regulating Monocyte/Macrophage Differentiation in
ApoE
−/−
Mice. Arterioscler Thromb Vasc Biol 2016; 36:491-500. [DOI: 10.1161/atvbaha.115.307057] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 01/12/2016] [Indexed: 01/17/2023]
Abstract
Objective—
Inflammatory monocytes/macrophages produce various proteinases, including matrix metalloproteinases, and degradation of the extracellular matrix by these activated proteinases weakens the mechanical strength of atherosclerotic plaques, which results in a rupture of the plaque. Peroxisome proliferator–activated receptor-γ induces a polarity shift of monocytes/macrophages toward less inflammatory phenotypes and has the potential to prevent atherosclerotic plaque ruptures. Therefore, we hypothesized that nanoparticle-mediated targeted delivery of the peroxisome proliferator–activated receptor-γ agonist pioglitazone into circulating monocytes could effectively inhibit plaque ruptures in a mouse model.
Approach and Results—
We prepared bioabsorbable poly(lactic-
co
-glycolic-acid) nanoparticles containing pioglitazone (pioglitazone-NPs). Intravenously administered poly(lactic-
co
-glycolic-acid) nanoparticles incorporated with fluorescein isothiocyanate were found in circulating monocytes and aortic macrophages by flow cytometric analysis. Weekly intravenous administration of pioglitazone-NPs (7 mg/kg per week) for 4 weeks decreased buried fibrous caps, a surrogate marker of plaque rupture, in the brachiocephalic arteries of
ApoE
−/−
mice fed a high-fat diet and infused with angiotensin II. In contrast, administration of control-NPs or an equivalent dose of oral pioglitazone treatment produced no effects. Pioglitazone-NPs inhibited the activity of matrix metalloproteinases and cathepsins in the brachiocephalic arteries. Pioglitazone-NPs regulated inflammatory cytokine expression and also suppressed the expression of extracellular matrix metalloproteinase inducer in bone marrow–derived macrophages.
Conclusions—
Nanoparticle-mediated delivery of pioglitazone inhibited macrophage activation and atherosclerotic plaque ruptures in hyperlipidemic
ApoE
−/−
mice. These results demonstrate a promising strategy with a favorable safety profile to prevent atherosclerotic plaque ruptures.
Collapse
Affiliation(s)
- Soichi Nakashiro
- From the Departments of Cardiovascular Medicine (S.N., T.M., R.U., J.K., M.T., S.K., K.S.) and Cardiovascular Research, Development, and Translational Medicine (K.N., K.E.), Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Tetsuya Matoba
- From the Departments of Cardiovascular Medicine (S.N., T.M., R.U., J.K., M.T., S.K., K.S.) and Cardiovascular Research, Development, and Translational Medicine (K.N., K.E.), Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Ryuta Umezu
- From the Departments of Cardiovascular Medicine (S.N., T.M., R.U., J.K., M.T., S.K., K.S.) and Cardiovascular Research, Development, and Translational Medicine (K.N., K.E.), Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Jun-ichiro Koga
- From the Departments of Cardiovascular Medicine (S.N., T.M., R.U., J.K., M.T., S.K., K.S.) and Cardiovascular Research, Development, and Translational Medicine (K.N., K.E.), Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Masaki Tokutome
- From the Departments of Cardiovascular Medicine (S.N., T.M., R.U., J.K., M.T., S.K., K.S.) and Cardiovascular Research, Development, and Translational Medicine (K.N., K.E.), Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Shunsuke Katsuki
- From the Departments of Cardiovascular Medicine (S.N., T.M., R.U., J.K., M.T., S.K., K.S.) and Cardiovascular Research, Development, and Translational Medicine (K.N., K.E.), Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kaku Nakano
- From the Departments of Cardiovascular Medicine (S.N., T.M., R.U., J.K., M.T., S.K., K.S.) and Cardiovascular Research, Development, and Translational Medicine (K.N., K.E.), Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kenji Sunagawa
- From the Departments of Cardiovascular Medicine (S.N., T.M., R.U., J.K., M.T., S.K., K.S.) and Cardiovascular Research, Development, and Translational Medicine (K.N., K.E.), Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kensuke Egashira
- From the Departments of Cardiovascular Medicine (S.N., T.M., R.U., J.K., M.T., S.K., K.S.) and Cardiovascular Research, Development, and Translational Medicine (K.N., K.E.), Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| |
Collapse
|
85
|
Lee SH, Choi JH. Involvement of Immune Cell Network in Aortic Valve Stenosis: Communication between Valvular Interstitial Cells and Immune Cells. Immune Netw 2016; 16:26-32. [PMID: 26937229 PMCID: PMC4770097 DOI: 10.4110/in.2016.16.1.26] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 01/14/2016] [Accepted: 01/16/2016] [Indexed: 01/11/2023] Open
Abstract
Aortic valve stenosis is a heart disease prevalent in the elderly characterized by valvular calcification, fibrosis, and inflammation, but its exact pathogenesis remains unclear. Previously, aortic valve stenosis was thought to be caused by chronic passive and degenerative changes associated with aging. However, recent studies have demonstrated that atherosclerotic processes and inflammation can induce valvular calcification and bone deposition, leading to valvular stenosis. In particular, the most abundant cell type in cardiac valves, valvular interstitial cells, can differentiate into myofibroblasts and osteoblast-like cells, leading to valvular calcification and stenosis. Differentiation of valvular interstitial cells can be trigged by inflammatory stimuli from several immune cell types, including macrophages, dendritic cells, T cells, B cells, and mast cells. This review indicates that crosstalk between immune cells and valvular interstitial cells plays an important role in the development of aortic valve stenosis.
Collapse
Affiliation(s)
- Seung Hyun Lee
- Department of Life Science, College of Natural Sciences, Research Institute of Natural Sciences, Hanyang University, Seoul 04763, Korea
| | - Jae-Hoon Choi
- Department of Life Science, College of Natural Sciences, Research Institute of Natural Sciences, Hanyang University, Seoul 04763, Korea
| |
Collapse
|
86
|
Zhang X, Jiang D, Li H. The interferon regulatory factors as novel potential targets in the treatment of cardiovascular diseases. Br J Pharmacol 2015; 172:5457-5476. [PMID: 25131895 PMCID: PMC4667854 DOI: 10.1111/bph.12881] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 07/26/2014] [Accepted: 08/12/2014] [Indexed: 02/06/2023] Open
Abstract
The family of interferon regulatory factors (IRFs) consists of nine members (IRF1-IRF9) in mammals. They act as transcription factors for the interferons and thus exert essential regulatory functions in the immune system and in oncogenesis. Recent clinical and experimental studies have identified critically important roles of the IRFs in cardiovascular diseases, arising from their participation in divergent and overlapping molecular programmes beyond the immune response. Here we review the current knowledge of the regulatory effects and mechanisms of IRFs on the immune system. The role of IRFs and their potential molecular mechanisms as novel stress sensors and mediators of cardiovascular diseases are highlighted.
Collapse
Affiliation(s)
- Xiao‐Jing Zhang
- Department of Cardiology, Renmin HospitalWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical SciencesUniversity of MacauMacaoChina
| | - Ding‐Sheng Jiang
- Department of Cardiology, Renmin HospitalWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
| | - Hongliang Li
- Department of Cardiology, Renmin HospitalWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
| |
Collapse
|
87
|
Kleinhenz JM, Murphy TC, Pokutta-Paskaleva AP, Gleason RL, Lyle AN, Taylor WR, Blount MA, Cheng J, Yang Q, Sutliff RL, Hart CM. Smooth Muscle-Targeted Overexpression of Peroxisome Proliferator Activated Receptor-γ Disrupts Vascular Wall Structure and Function. PLoS One 2015; 10:e0139756. [PMID: 26451838 PMCID: PMC4599849 DOI: 10.1371/journal.pone.0139756] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 09/15/2015] [Indexed: 12/15/2022] Open
Abstract
Activation of the nuclear hormone receptor, PPARγ, with pharmacological agonists promotes a contractile vascular smooth muscle cell phenotype and reduces oxidative stress and cell proliferation, particularly under pathological conditions including vascular injury, restenosis, and atherosclerosis. However, pharmacological agonists activate both PPARγ-dependent and -independent mechanisms in multiple cell types confounding efforts to clarify the precise role of PPARγ in smooth muscle cell structure and function in vivo. We, therefore, designed and characterized a mouse model with smooth muscle cell-targeted PPARγ overexpression (smPPARγOE). Our results demonstrate that smPPARγOE attenuated contractile responses in aortic rings, increased aortic compliance, caused aortic dilatation, and reduced mean arterial pressure. Molecular characterization revealed that compared to littermate control mice, aortas from smPPARγOE mice expressed lower levels of contractile proteins and increased levels of adipocyte-specific transcripts. Morphological analysis demonstrated increased lipid deposition in the vascular media and in smooth muscle of extravascular tissues. In vitro adenoviral-mediated PPARγ overexpression in human aortic smooth muscle cells similarly increased adipocyte markers and lipid uptake. The findings demonstrate that smooth muscle PPARγ overexpression disrupts vascular wall structure and function, emphasizing that balanced PPARγ activity is essential for vascular smooth muscle homeostasis.
Collapse
Affiliation(s)
- Jennifer M. Kleinhenz
- Atlanta VA Medical Center, Decatur, GA, United States of America
- Emory University, Atlanta, GA, United States of America
| | - Tamara C. Murphy
- Atlanta VA Medical Center, Decatur, GA, United States of America
- Emory University, Atlanta, GA, United States of America
| | | | | | | | - W. Robert Taylor
- Atlanta VA Medical Center, Decatur, GA, United States of America
- Emory University, Atlanta, GA, United States of America
- Georgia Institute of Technology, Atlanta, GA, United States of America
| | | | - Juan Cheng
- Emory University, Atlanta, GA, United States of America
| | - Qinglin Yang
- University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Roy L. Sutliff
- Atlanta VA Medical Center, Decatur, GA, United States of America
- Emory University, Atlanta, GA, United States of America
| | - C. Michael Hart
- Atlanta VA Medical Center, Decatur, GA, United States of America
- Emory University, Atlanta, GA, United States of America
- * E-mail:
| |
Collapse
|
88
|
Tsukahara T, Tsukahara R, Haniu H, Matsuda Y, Murakami-Murofushi K. Cyclic phosphatidic acid inhibits the secretion of vascular endothelial growth factor from diabetic human coronary artery endothelial cells through peroxisome proliferator-activated receptor gamma. Mol Cell Endocrinol 2015; 412:320-9. [PMID: 26007326 DOI: 10.1016/j.mce.2015.05.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 05/16/2015] [Accepted: 05/18/2015] [Indexed: 12/27/2022]
Abstract
Atherosclerosis is a disease characterized by building up plaques formation and leads to a potentially serious condition in which arteries are clogged by fatty substances such as cholesterol. Increasing evidence suggests that atherosclerosis is accelerated in type 2 diabetes. Recent study reported that high level of alkyl glycerophosphate (AGP) was accumulated in atherosclerotic lesions. The presence of this phospholipid in mildly oxidized low-density lipoprotein (LDL) is likely to be involved in atherogenesis. It has been reported that the activation of peroxisome proliferator-activated receptor gamma plays a key role in developing atherosclerosis. Our previous result indicates that cyclic phosphatidic acid (cPA), one of bioactive lipids, potently suppresses neointima formation by inhibiting the activation of peroxisome proliferator-activated receptor gamma (PPARγ). However, the detailed mechanism is still unclear. In this study, to elucidate the mechanism of the cPA-PPARγ axis in the coronary artery endothelium, especially in patients with type 2 diabetes, we investigated the proliferation, migration, and secretion of VEGF in human coronary artery endothelial cells from diabetes patients (D-HCAECs). AGP induced cell growth and migration; however, cPA suppressed the AGP-elicited growth and migration in D-HCAECs. Moreover, AGP increased VEGF secretion from D-HCAECs, and this event was attenuated by cPA. Taken together, these results suggest that cPA suppresses VEGF-stimulated growth and migration in D-HCAECs. These findings could be important for regulatory roles of PPARγ and VEGF in the vascular processes associated with diabetes and atherosclerosis.
Collapse
Affiliation(s)
- Tamotsu Tsukahara
- Department of Molecular Pharmacology and Neuroscience, Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan.
| | - Ryoko Tsukahara
- Endowed Research Division of Human Welfare Sciences, Ochanomizu University, 2-1-1, Ohtsuka, Bunkyo-ku, Tokyo 112-8610, Japan; Science and Education Center, Ochanomizu University, 2-1-1 Ohtsuka, Bunkyo-ku, Tokyo 112-861, Japan
| | - Hisao Haniu
- Institue for Biomedical Sciences, Shinshu University Interdisciplinary Cluster for Cutting Edge Research 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Yoshikazu Matsuda
- Clinical Pharmacology Educational Center, Nihon Pharmaceutical University, Ina-machi, Saitama 362-0806, Japan
| | | |
Collapse
|
89
|
Tiyerili V, Becher UM, Camara B, Yildirimtürk C, Aksoy A, Kebschull M, Werner N, Nickenig G, Müller C. Impact of peroxisome proliferator-activated receptor γ on angiotensin II type 1 receptor-mediated insulin sensitivity, vascular inflammation and atherogenesis in hypercholesterolemic mice. Arch Med Sci 2015; 11:877-85. [PMID: 26322101 PMCID: PMC4548041 DOI: 10.5114/aoms.2015.53309] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 08/05/2013] [Accepted: 08/14/2013] [Indexed: 01/13/2023] Open
Abstract
INTRODUCTION The angiotensin II type 1 receptor (AT1R) and the peroxisome proliferator-activated receptor γ (PPARγ) have been implicated in the pathogenesis of atherosclerosis. A number of studies have reported that AT1R inhibition or genetic AT1R disruption and PPARγ activation inhibit vascular inflammation and improve glucose and lipid metabolism, underscoring a molecular interaction of AT1R and PPARγ. We here analyzed the hypothesis that vasculoprotective anti-inflammatory and metabolic effects of AT1R inhibition are mediated by PPARγ. MATERIAL AND METHODS Female ApoE(-/-)/AT1R(-/-) mice were fedwith a high-fat and cholesterol-rich diet and received continuous treatment with the selective PPARγ antagonist GW9662 or vehicle at a rate of 700 ng/kg/min for 4 weeks using subcutaneously implanted osmotic mini-pumps. Additionally, one group of female ApoE(-/-) mice served as a control group. After treatment for 4 weeks mice were sacrificed and read-outs (plaque development, vascular inflammation and insulinsensitivity) were performed. RESULTS Using AT1R deficient ApoE(-/-) mice (ApoE(-/-)/AT1R(-/-) mice) we found decreased cholesterol-induced endothelial dysfunction and atherogenesis compared to ApoE(-/-) mice. Inhibition of PPARγ by application of the specific PPARγ antagonist GW9662 significantly abolished the anti-atherogenic effects of AT1R deficiency in ApoE(-/-)/AT1R(-/-) mice (plaque area as % of control: ApoE(-/-): 39 ±5%; ApoE(-/-)/AT1R(-/-): 17 ±7%, p = 0.044 vs. ApoE(-/-); ApoE(-/-)/AT1R(-/-) + GW9662: 31 ±8%, p = 0.047 vs. ApoE(-/-)/AT1R(-/-)). Focusing on IL6 as a pro-inflammatory humoral marker we detected significantly increased IL-6 levels in GW9662-treated animals (IL-6 in pg/ml: ApoE(-/-): 230 ±16; ApoE(-/-)/AT1R(-/-): 117 ±20, p = 0.01 vs. ApoE(-/-); ApoE(-/-)/AT1R(-/-) + GW9662: 199 ±20, p = 0.01 vs. ApoE(-/-)/AT1R(-/-)), while the anti-inflammatory marker IL-10 was significantly reduced after PPARγ inhibition in GW9662 animals (IL-10 in pg/ml: ApoE(-/-): 18 ±4; ApoE(-/-)/AT1R(-/-): 55 ±12, p = 0.03 vs. ApoE(-/-); ApoE(-/-)/AT1R(-/-) + GW9662: 19 ±4, p = 0.03 vs. ApoE(-/-)/AT1R(-/-)). Metabolic parameters of glucose homeostasis (glucose and insulin tolerance test) were significantly deteriorated in ApoE(-/-)/AT1R(-/-) mice treated with GW9662 as compared to vehicle-treated ApoE(-/-)/AT1R(-/-) mice. Systolic blood pressure and plasma cholesterol levels were similar in all groups. CONCLUSIONS Genetic disruption of the AT1R attenuates atherosclerosis and improves endothelial function in an ApoE(-/-) mouse model of hypercholesterolemia-induced atherosclerosis via PPARγ, indicating a significant role of PPARγ in reduced vascular inflammation, improvement of insulin sensitivity and atheroprotection of AT1R deficiency.
Collapse
Affiliation(s)
- Vedat Tiyerili
- Medizinische Klinik und Poliklinik II, Innere Medizin, Universitätsklinikum Bonn, Bonn, Germany
| | - Ulrich M Becher
- Medizinische Klinik und Poliklinik II, Innere Medizin, Universitätsklinikum Bonn, Bonn, Germany
| | - Bakary Camara
- Medizinische Klinik und Poliklinik II, Innere Medizin, Universitätsklinikum Bonn, Bonn, Germany
| | - Cihan Yildirimtürk
- Medizinische Klinik und Poliklinik II, Innere Medizin, Universitätsklinikum Bonn, Bonn, Germany
| | - Adem Aksoy
- Medizinische Klinik und Poliklinik II, Innere Medizin, Universitätsklinikum Bonn, Bonn, Germany
| | - Moritz Kebschull
- Medizinische Klinik und Poliklinik II, Innere Medizin, Universitätsklinikum Bonn, Bonn, Germany
| | - Nikos Werner
- Medizinische Klinik und Poliklinik II, Innere Medizin, Universitätsklinikum Bonn, Bonn, Germany
| | - Georg Nickenig
- Medizinische Klinik und Poliklinik II, Innere Medizin, Universitätsklinikum Bonn, Bonn, Germany
| | - Cornelius Müller
- Medizinische Klinik und Poliklinik II, Innere Medizin, Universitätsklinikum Bonn, Bonn, Germany
| |
Collapse
|
90
|
Rosiglitazone impedes Porphyromonas gingivalis-accelerated atherosclerosis by downregulating the TLR/NF-κB signaling pathway in atherosclerotic mice. Int Immunopharmacol 2015; 23:701-8. [PMID: 25445963 DOI: 10.1016/j.intimp.2014.10.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 10/22/2014] [Accepted: 10/22/2014] [Indexed: 11/20/2022]
Abstract
Porphyromonas gingivalis,a predominant periodontal pathogen, is known to accelerate atherosclerosis in hyperlipidemic animals via aberrant inflammatory responses. Peroxisome proliferator-activated receptor gamma (PPARγ) agonists have been reported to exert anti-inflammatory effects in vitro. The purpose of the present study was to investigate the potential protective role of the PPARγ agonist rosiglitazone in pathogen accelerated atherosclerosis in an apolipoprotein E-deficient (ApoE-/-) mouse model. ApoE-/- mice were inoculated intravenously with live P. gingivalis (strain 33277) or the buffer vehicle and treated with rosiglitazone or saline over a 10-week period. Their atherosclerotic status in aortic artery was assessed through histomorphometric analysis, inflammatory agent and lipid profiles in blood was determined by ELISA, and levels of relevant cytokines and Toll-like receptors (TLRs) in aortic tissues were evaluated using immunohistochemistry and quantitative PCR. P. gingivalis inoculation was associated with increased atherosclerotic plaque formation in the aorta and higher levels of serum pro-inflammatory cytokines (tumor necrosis factor-α, monocyte chemotactic protein-1 and interleukin-1β), but the serum lipid profile was not affected by P. gingivalis infection. Levels of tumor necrosis factor-α, monocyte chemotactic protein-1 intercellular cell adhesion molecule-1 and TLRs were higher in the aortic tissues of mice exposed to P. gingivalis, and activation of nuclear factor-κB was also observed. In both P. gingivalis-treated and -untreated ApoE-/- mice, rosiglitazone treatment was associated with less atherosclerotic plaque formation; lower serum inflammatory cytokines, total cholesterol, and low density lipoprotein cholesterol; higher levels of PPARγ, lower amounts of TLR2/4 and downregulated nuclear factor-κB activity in aortic tissues. These findings suggest that rosiglitazone mitigates or prevents P. gingivalis-accelerated atherosclerosis by inhibiting the inflammatory response via downregulation of the TLR/ nuclear factor-κB signaling pathway.
Collapse
|
91
|
Abstract
Early brain injury (EBI) plays a crucial role in the pathological progress of subarachnoid hemorrhage (SAH). This study was designed to determine whether rosiglitazone protects the brain against EBI in rats, and discuss the role of the anti-apoptotic mechanism mediated by Bcl-2 family proteins in this neuroprotection. 86 male Sprague-Dawley rats were divided into the sham group, the SAH+ vehicle group and the SAH+ rosiglitazone group. SAH was induced via an endovascular perforation technique and rosiglitazone (3mg/kg) or vehicle was administered. Mortality, neurological scores, brain water content, Evans blue dye assay, TUNEL stain assay, Gelatin zymography, and western blot analysis were performed. Rosiglitazone significantly improved mortality, neurological scores, brain water content, blood brain barrier (BBB) and apoptosis compared with the vehicle group within 24h after SAH. The TUNEL staining assay demonstrated that apoptosis was ameliorated. Cleaved Caspase-3 and MMP-9 expression was reduced, whereas Bcl-2 and p-Bad was markedly preserved by rosiglitazone. A significant elevation of p-Akt was detected after rosiglitazone treatment. Our study demonstrated that rosiglitazone plays a neuroprotective role in EBI after SAH via attenuation of BBB disruption, brain edema and apoptosis.
Collapse
|
92
|
Propofol up-regulates expression of ABCA1, ABCG1, and SR-B1 through the PPARγ/LXRα signaling pathway in THP-1 macrophage-derived foam cells. Cardiovasc Pathol 2015; 24:230-5. [DOI: 10.1016/j.carpath.2014.12.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 12/02/2014] [Accepted: 12/19/2014] [Indexed: 11/18/2022] Open
|
93
|
Oe H, Nakamura K, Kihara H, Shimada K, Fukuda S, Takagi T, Miyoshi T, Hirata K, Yoshikawa J, Ito H. Comparison of effects of sitagliptin and voglibose on left ventricular diastolic dysfunction in patients with type 2 diabetes: results of the 3D trial. Cardiovasc Diabetol 2015; 14:83. [PMID: 26084668 PMCID: PMC4473835 DOI: 10.1186/s12933-015-0242-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 06/05/2015] [Indexed: 01/20/2023] Open
Abstract
Background Left ventricular (LV) diastolic dysfunction is frequently observed in patients with type 2 diabetes. Dipeptidyl peptidase-4 inhibitor (DPP-4i) attenuates postprandial hyperglycemia (PPH) and may have cardio-protective effects. It remains unclear whether DPP-4i improves LV diastolic function in patients with type 2 diabetes, and, if so, it is attributable to the attenuation of PPH or to a direct cardiac effect of DPP-4i. We compared the effects of the DPP-4i, sitagliptin, and the alpha-glucosidase inhibitor, voglibose, on LV diastolic function in patients with type 2 diabetes. Methods We conducted a prospective, randomized, open-label, multicenter study of 100 diabetic patients with LV diastolic dysfunction. Patients received sitagliptin (50 mg/day) or voglibose (0.6 mg/day). The primary endpoints were changes in the e’ velocity and E/e’ ratio from baseline to 24 weeks later. The secondary efficacy measures included HbA1c, GLP-1, lipid profiles, oxidative stress markers and inflammatory markers. Results The study was completed with 40 patients in the sitagliptin group and 40 patients in the voglibose group. There were no significant changes in the e’ velocity and E/e’ ratio from baseline to 24 weeks later in both groups. However, analysis of covariance demonstrated that pioglitazone use is an independent factor associated with changes in the e’ and E/e’ ratio. Among patients not using pioglitazone, e’ increased and the E/e’ ratio decreased in both the sitagliptin and voglibose groups. GLP-1 level increased from baseline to 24 weeks later only in the sitagliptin group (4.8 ± 4.7 vs. 7.3 ± 5.5 pmol/L, p < 0.05). The reductions in HbA1c and body weight were significantly greater in the sitagliptin group than in the voglibose group (−0.7 ± 0.6 % vs. −0.3 ± 0.4, p < 0.005; −1.3 ± 3.2 kg vs. 0.4 ± 2.8 kg, p < 0.05, respectively). There were no changes in lipid profiles and inflammatory markers in both groups. Conclusions Our trial showed that sitagliptin reduces HbA1c levels more greatly than voglibose does, but that neither was associated with improvement in the echocardiographic parameters of LV diastolic function in patients with diabetes. Trial registration Registered at http://www.umin.ac.jp under UMIN000003784
Collapse
Affiliation(s)
- Hiroki Oe
- Center of Ultrasonic Diagnostics, Okayama University Hospital, Okayama, Japan.
| | - Kazufumi Nakamura
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan.
| | - Hajime Kihara
- Department of Internal Medicine, Kihara Cardiovascular Clinic, Asahikawa, Japan.
| | - Kenei Shimada
- Department of Internal Medicine and Cardiology, Osaka City University of Medicine, Osaka, Japan.
| | - Shota Fukuda
- Department of Medicine, Osaka Ekisaikai Hospital, Osaka, Japan.
| | | | - Toru Miyoshi
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan.
| | - Kumiko Hirata
- Department of Cardiovascular Medicine, Wakayama Medical University, Wakayama, Japan.
| | | | - Hiroshi Ito
- Department of Cardiovascular Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan.
| | | |
Collapse
|
94
|
Lin BF, Kuo CY, Wen LL, Chen CM, Chang YF, Wong CS, Cherng CH, Chuang MY, Wu ZF. Rosiglitazone attenuates cerebral vasospasm and provides neuroprotection in an experimental rat model of subarachnoid hemorrhage. Neurocrit Care 2015; 21:316-31. [PMID: 25022803 DOI: 10.1007/s12028-014-0010-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Glutamate and oxidative stress play important roles after subarachnoid hemorrhage (SAH). The ability to modulate glutamate transporter 1 (GLT-1) and the antioxidative effect of rosiglitazone have been demonstrated. We investigated the neuroprotective effect of rosiglitazone after SAH. METHODS SAH was induced by double blood injection. The rats were randomly divided into sham, SAH + vehicle, and SAH + rosiglitazone groups and treated with dimethyl sulfoxide, dimethyl sulfoxide, and 6 mg/kg of rosiglitazone, respectively, at 2 and 12 h after SAH induction and then daily for 6 days. Cerebrospinal fluid dialysates were collected 30 min before SAH induction and then daily for 7 days for glutamate measurement. Mortality, body weight, and neurological scores were also measured daily. On day 7 after SAH, the wall thickness and the perimeter of the basilar artery (BA), neuron variability, GLT-1 levels, glial fibrillary acidic protein (GFAP) expression and activity, and malondialdehyde, superoxide dismutase, and catalase activities were also evaluated. RESULTS Rosiglitazone improved survival (relative risk = 0.325) and neurological functions and reduced neuronal degeneration (5.7 ± 0.8 vs. 10.0 ± 0.9; P < 0.001) compared with the SAH + vehicle group. Rosiglitazone also lowered glutamate levels by 43.5-fold and upregulated GLT-1 expression by 1.5-fold and astrocyte activity by 1.8-fold compared with the SAH + vehicle group. The increase in BA wall thickness was significantly attenuated by rosiglitazone, whereas the perimeter of the BA was increased. In addition, rosiglitazone abated the 1.9-fold increase in malondialdehyde levels and the 1.6-fold increase in catalase activity after SAH. CONCLUSION Rosiglitazone reduced SAH mortality, neurological deficits, body weight loss, GFAP loss, and cerebral vasospasm by preventing the neurotoxicity induced by glutamate and oxidative stress.
Collapse
Affiliation(s)
- Bo-Feng Lin
- Department of Anesthesiology, Tri-Service General Hospital, National Defense Medical Center, #325, Section 2 Chenggung Road, Neihu 114, Taipei, Taiwan, ROC,
| | | | | | | | | | | | | | | | | |
Collapse
|
95
|
PGC-1α overexpression suppresses blood pressure elevation in DOCA-salt hypertensive mice. Biosci Rep 2015; 35:BSR20150076. [PMID: 26182379 PMCID: PMC4613682 DOI: 10.1042/bsr20150076] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 05/15/2015] [Indexed: 02/02/2023] Open
Abstract
Increasing evidences have accumulated that endothelial dysfunction is involved in the pathogenesis of hypertension. Peroxisome proliferator-activated receptor γ (PPARγ) coactivator-1α (PGC-1α) has been identified as an essential factor that protects against endothelial dysfunction in vascular pathologies. However, the functional role of PGC-1α in hypertension is not well understood. Using an adenovirus infection model, we tested the hypothesis that PGC-1α overexpression retards the progression of hypertension in deoxycorticosterone acetate (DOCA)-salt mice model through preservation of the function of endothelium. We first demonstrated that PGC-1α expression not only in conductance and resistance arteries but also in endothelial cells was decreased after DOCA-salt treatment. In PGC-1α adenovirus-infected mice, the elevation of blood pressure in DOCA-salt mice was attenuated, as determined using tail-cuff measurement. Furthermore, PGC-1α overexpression inhibited the decrease in nitric oxide (NO) generation and the increase in superoxide anion (O2 (-)) production in DOCA-salt-treated mice, in parallel with improved endothelium-dependent relaxation. Rather than affecting endothelial NO synthase (eNOS) total expression and phosphorylation, PGC-1α significantly inhibited eNOS uncoupling, as evidenced by increased eNOS homodimerization, BH4 levels, GTP-cyclohydrolase 1 (GTPCH1) and dihydrofolate reductase (DHFR) expression and heat-shock protein (Hsp)90-eNOS interaction. Our findings demonstrate that PGC-1α overexpression preserves eNOS coupling, enhances NO generation, improves endothelium-dependent relaxation and thus lowers blood pressure, suggesting that up-regulation of PGC-1α may be a novel strategy to prevent and treat hypertension.
Collapse
|
96
|
Abernethy AD, Stackhouse K, Hart S, Devendra G, Bashore TM, Dweik R, Krasuski RA. Impact of diabetes in patients with pulmonary hypertension. Pulm Circ 2015; 5:117-23. [PMID: 25992276 DOI: 10.1086/679705] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 08/26/2014] [Indexed: 12/20/2022] Open
Abstract
Diabetes complicates management in a number of disease states and adversely impacts survival; how diabetes affects patients with pulmonary hypertension (PH) has not been well characterized. With insulin resistance having recently been demonstrated in PH, we sought to examine the impact of diabetes in these patients. Demographic characteristics, echo data, and invasive hemodynamic data were prospectively collected for 261 patients with PH referred for initial hemodynamic assessment. Diabetes was defined as documented insulin resistance or treatment with antidiabetic medications. Fifty-five patients (21%) had diabetes, and compared with nondiabetic patients, they were older (mean years ± SD, 61 ± 13 vs. 56 ± 16; [Formula: see text]), more likely to be black (29% vs. 14%; [Formula: see text]) and hypertensive (71% vs. 30%; [Formula: see text]), and had higher mean (±SD) serum creatinine levels (1.1 ± 0.5 vs. 1.0 ± 0.4; [Formula: see text]). Diabetic patients had similar World Health Organization functional class at presentation but were more likely to have pulmonary venous etiology of PH (24% vs. 10%; [Formula: see text]). Echo findings, including biventricular function, tricuspid regurgitation, and pressure estimates were similar. Invasive pulmonary pressures and cardiac output were similar, but right atrial pressure was appreciably higher (14 ± 8 mmHg vs. 10 ± 5 mmHg; [Formula: see text]). Despite similar management, survival was markedly worse and remained so after statistical adjustment. In summary, diabetic patients referred for assessment of PH were more likely to have pulmonary venous disease than nondiabetic patients with PH, with hemodynamics suggesting greater right-sided diastolic dysfunction. The markedly worse survival in these patients merits further study.
Collapse
Affiliation(s)
- Abraham D Abernethy
- Department of Internal Medicine/Pediatrics, University Hospitals Case Medical Center, Cleveland, Ohio, USA
| | - Kathryn Stackhouse
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
| | - Stephen Hart
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
| | - Ganesh Devendra
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
| | - Thomas M Bashore
- Department of Cardiovascular Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Raed Dweik
- Department of Pulmonary Medicine, Cleveland Clinic Respiratory Institute, Cleveland, Ohio, USA
| | - Richard A Krasuski
- Department of Cardiovascular Medicine, Cleveland Clinic Heart and Vascular Institute, Cleveland, Ohio, USA
| |
Collapse
|
97
|
Atkins GB, Orasanu G, Jain MK. Endothelial Cells. Atherosclerosis 2015. [DOI: 10.1002/9781118828533.ch9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
98
|
Jin H, Gebska MA, Blokhin IO, Wilson KM, Ketsawatsomkron P, Chauhan AK, Keen HL, Sigmund CD, Lentz SR. Endothelial PPAR-γ protects against vascular thrombosis by downregulating P-selectin expression. Arterioscler Thromb Vasc Biol 2015; 35:838-44. [PMID: 25675995 DOI: 10.1161/atvbaha.115.305378] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE We tested the hypothesis that endothelial peroxisome proliferator-activated receptor-γ protects against vascular thrombosis using a transgenic mouse model expressing a peroxisome proliferator-activated receptor-γ mutant (E-V290M) selectively in endothelium. APPROACH AND RESULTS The time to occlusive thrombosis of the carotid artery was significantly shortened in E-V290M mice compared with nontransgenic littermates after either chemical injury with ferric chloride (5.1 ± 0.2 versus 10.1 ± 3.3 minutes; P=0.01) or photochemical injury with rose bengal (48 ± 9 versus 74 ± 9 minutes; P=0.04). Gene set enrichment analysis demonstrated the upregulation of NF-κB target genes, including P-selectin, in aortic endothelial cells from E-V290M mice (P<0.001). Plasma P-selectin and carotid artery P-selectin mRNA were elevated in E-V290M mice (P<0.05). P-selectin-dependent leukocyte rolling on mesenteric venules was increased in E-V290M mice compared with nontransgenic mice (53 ± 8 versus 25 ± 7 per minute; P=0.02). The shortened time to arterial occlusion in E-V290M mice was reversed by administration of P-selectin-blocking antibodies or neutrophil-depleting antibodies (P=0.04 and P=0.02, respectively) before photochemical injury. CONCLUSIONS Endothelial peroxisome proliferator-activated receptor-γ protects against thrombosis through a mechanism that involves downregulation of P-selectin expression and diminished P-selectin-mediated leukocyte-endothelial interactions.
Collapse
Affiliation(s)
- Hong Jin
- From the Departments of Internal Medicine (H.J., M.A.G., I.O.B., K.M.W., A.K.C., C.D.S., S.R.L.) and Pharmacology (P.K., H.L.K., C.D.S.), Interdisciplinary Graduate Program in Molecular and Cellular Biology (I.O.B.), University of Iowa Carver College of Medicine, Iowa City
| | - Milena A Gebska
- From the Departments of Internal Medicine (H.J., M.A.G., I.O.B., K.M.W., A.K.C., C.D.S., S.R.L.) and Pharmacology (P.K., H.L.K., C.D.S.), Interdisciplinary Graduate Program in Molecular and Cellular Biology (I.O.B.), University of Iowa Carver College of Medicine, Iowa City
| | - Ilya O Blokhin
- From the Departments of Internal Medicine (H.J., M.A.G., I.O.B., K.M.W., A.K.C., C.D.S., S.R.L.) and Pharmacology (P.K., H.L.K., C.D.S.), Interdisciplinary Graduate Program in Molecular and Cellular Biology (I.O.B.), University of Iowa Carver College of Medicine, Iowa City
| | - Katina M Wilson
- From the Departments of Internal Medicine (H.J., M.A.G., I.O.B., K.M.W., A.K.C., C.D.S., S.R.L.) and Pharmacology (P.K., H.L.K., C.D.S.), Interdisciplinary Graduate Program in Molecular and Cellular Biology (I.O.B.), University of Iowa Carver College of Medicine, Iowa City
| | - Pimonrat Ketsawatsomkron
- From the Departments of Internal Medicine (H.J., M.A.G., I.O.B., K.M.W., A.K.C., C.D.S., S.R.L.) and Pharmacology (P.K., H.L.K., C.D.S.), Interdisciplinary Graduate Program in Molecular and Cellular Biology (I.O.B.), University of Iowa Carver College of Medicine, Iowa City
| | - Anil K Chauhan
- From the Departments of Internal Medicine (H.J., M.A.G., I.O.B., K.M.W., A.K.C., C.D.S., S.R.L.) and Pharmacology (P.K., H.L.K., C.D.S.), Interdisciplinary Graduate Program in Molecular and Cellular Biology (I.O.B.), University of Iowa Carver College of Medicine, Iowa City
| | - Henry L Keen
- From the Departments of Internal Medicine (H.J., M.A.G., I.O.B., K.M.W., A.K.C., C.D.S., S.R.L.) and Pharmacology (P.K., H.L.K., C.D.S.), Interdisciplinary Graduate Program in Molecular and Cellular Biology (I.O.B.), University of Iowa Carver College of Medicine, Iowa City
| | - Curt D Sigmund
- From the Departments of Internal Medicine (H.J., M.A.G., I.O.B., K.M.W., A.K.C., C.D.S., S.R.L.) and Pharmacology (P.K., H.L.K., C.D.S.), Interdisciplinary Graduate Program in Molecular and Cellular Biology (I.O.B.), University of Iowa Carver College of Medicine, Iowa City
| | - Steven R Lentz
- From the Departments of Internal Medicine (H.J., M.A.G., I.O.B., K.M.W., A.K.C., C.D.S., S.R.L.) and Pharmacology (P.K., H.L.K., C.D.S.), Interdisciplinary Graduate Program in Molecular and Cellular Biology (I.O.B.), University of Iowa Carver College of Medicine, Iowa City.
| |
Collapse
|
99
|
Liu W, Li J, Liu M, Zhang H, Wang N. PPAR-γ Promotes Endothelial Cell Migration By Inducing the Expression of Sema3g. J Cell Biochem 2015; 116:514-23. [DOI: 10.1002/jcb.24994] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 10/14/2014] [Indexed: 01/01/2023]
Affiliation(s)
- Weiwei Liu
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences; Ministry of Education; Peking University Health Science Center; Beijing China
| | - Jingjin Li
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences; Ministry of Education; Peking University Health Science Center; Beijing China
- Department of Cardiology; Peking University People's Hospital; Beijing China
| | - Min Liu
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences; Ministry of Education; Peking University Health Science Center; Beijing China
| | - Hong Zhang
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences; Ministry of Education; Peking University Health Science Center; Beijing China
| | - Nanping Wang
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences; Ministry of Education; Peking University Health Science Center; Beijing China
| |
Collapse
|
100
|
Pioglitazone, a peroxisome proliferator-activated receptor γ activator, suppresses coronary spasm. Coron Artery Dis 2014; 25:671-7. [DOI: 10.1097/mca.0000000000000144] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|