51
|
Hoppe C, Jacob E, Styles L, Kuypers F, Larkin S, Vichinsky E. Simvastatin reduces vaso-occlusive pain in sickle cell anaemia: a pilot efficacy trial. Br J Haematol 2017; 177:620-629. [PMID: 28369718 PMCID: PMC5435522 DOI: 10.1111/bjh.14580] [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: 10/05/2016] [Accepted: 12/06/2016] [Indexed: 12/21/2022]
Abstract
Sickle cell anaemia (SCA) is a progressive vascular disease characterized by episodic vaso-occlusive pain. Despite the broad impact of inflammation on acute and chronic clinical manifestations of SCA, no directed anti-inflammatory therapies currently exist. Statins are cholesterol-lowering agents shown to confer protection from vascular injury by suppressing inflammation. We previously documented a reduction in soluble biomarkers of inflammation in patients with sickle cell disease treated with simvastatin. To determine the potential clinical efficacy of simvastatin, we treated 19 SCA patients with single daily dose simvastatin for 3 months and assessed changes from baseline in the frequency and intensity of diary-reported pain and levels of circulating nitric oxide metabolites (NOx), high sensitivity C-reactive protein (hs-CRP), vascular cell adhesion molecule 1 (VCAM-1), intercellular adhesion molecule 1 (ICAM-1), ICAM-3, E-selectin, and vascular endothelial growth factor (VEGF). Treatment with simvastatin resulted in a significant reduction in the frequency of pain (P = 0·0003), oral analgesic use (P = 0·003) and circulating hs-CRP (P = 0·003), soluble (s)E-selectin (P = 0·01), sICAM-1 (P = 0·02), sICAM-3 (P = 0·02) and sVEGF (P = 0·01). Simvastatin had no effect on pain intensity or levels of NOx, sP-selectin and sVCAM-1. The observed reductions in pain rate and markers of inflammation were greatest in subjects receiving hydroxycarbamide (HC), suggesting a synergistic effect of simvastatin. These results provide preliminary clinical data to support a larger trial of simvastatin in SCA.
Collapse
Affiliation(s)
- Carolyn Hoppe
- Department of Hematology-Oncology, UCSF Benioff Children’s Hospital Oakland, Oakland, CA
| | - Eufemia Jacob
- School of Nursing, University of California Los Angeles, CA
| | | | - Frans Kuypers
- Children’s Hospital Oakland Research Institute, Oakland, CA
| | - Sandra Larkin
- Children’s Hospital Oakland Research Institute, Oakland, CA
| | - Elliott Vichinsky
- Department of Hematology-Oncology, UCSF Benioff Children’s Hospital Oakland, Oakland, CA
| |
Collapse
|
52
|
Li Y, Lu G, Sun D, Zuo H, Wang DW, Yan J. Inhibition of endoplasmic reticulum stress signaling pathway: A new mechanism of statins to suppress the development of abdominal aortic aneurysm. PLoS One 2017; 12:e0174821. [PMID: 28369137 PMCID: PMC5378361 DOI: 10.1371/journal.pone.0174821] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 03/15/2017] [Indexed: 12/02/2022] Open
Abstract
Background Abdominal aortic aneurysm (AAA) is a potentially lethal disease with extremely poor survival rates once the aneurysm ruptures. Statins may exert beneficial effects on the progression of AAA. However, the underlying mechanism is still not known. The purpose of the present study is to investigate whether statin could inhibit AAA formation by inhibiting the endoplasmic reticulum (ER) stress signal pathway. Methods A clinically relevant AAA model was induced in Apolipoprotein E-deficient (ApoE−/−) mice, which were infused with angiotensin II (Ang II) for 28 days. These mice were randomly divided into following 4 groups: saline infusion alone; Ang II infusion alone; Ang II infusion plus Atorvastatin (20mg/kg/d); and Ang II infusion plus Atorvastatin (30mg/kg/d). Besides, another AAA model was induced in C57 mice with extraluminal CaCl2, which were divided into 3 groups: sham group, CaCl2-induced AAA group, and CaCl2-induced AAA plus atorvastatin (20mg/kg/d) group. Then, aortic tissue was excised for further examinations, respectively. In vitro studies, Ang II with or without simvastatin treatment were applied to the vascular smooth muscle cells (VSMCS) and Raw 264.7 cells. The ER stress signal pathway, apoptosis and inflammatory response were evaluated by in vivo and in vitro assays. Results We found that higher dose of atorvastatin can effectively suppress the development and progression of AAA induced by Ang II or CaCl2. Mechanistically, the activation of ER stress and inflammatory response were found involved in Ang II-induced AAA formation. The atorvastatin infusion significantly reduced ER stress signaling proteins, the number of apoptotic cells, and the activation of Caspase12 and Bax in the Ang II-induced ApoE−/− mice, compared with mice treated by Ang II alone. Furthermore, proinflammatory cytokines such as IL-6, IL-8, IL-1β were all remarkably inhibited after atorvastatin treatment. In vitro, the inhibitory effect of simvastatin on the ER stress signal pathway could be observed in both vascular smooth muscle cells and macrophages, and these inhibitory effects of statin were in a dose-dependent manner. In addition, apoptosis was induced with Ang II treatment. The maximal inhibitory effect of simvastatin on apoptosis was observed at 10 μmol/l. Conclusions We conclude that higher dose of statin can effectively suppress the development of AAA, and reduce ER stress, ER stress-associated apoptosis signaling pathways, and inflammatory response. These findings reveal a new mechanism underlying the inhibitory effect of statin on AAA formation/progression.
Collapse
MESH Headings
- Angiotensin II
- Animals
- Aorta/drug effects
- Aorta/metabolism
- Aorta/pathology
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/prevention & control
- Apolipoproteins E/deficiency
- Apolipoproteins E/genetics
- Apoptosis/drug effects
- Apoptosis/physiology
- Atorvastatin/pharmacology
- Calcium Chloride
- Cell Line
- Cytokines/metabolism
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Endoplasmic Reticulum Stress/drug effects
- Endoplasmic Reticulum Stress/physiology
- Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology
- Macrophages/drug effects
- Macrophages/metabolism
- Macrophages/pathology
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Random Allocation
Collapse
Affiliation(s)
- Yuanyuan Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gangsheng Lu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dating Sun
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Houjuan Zuo
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- * E-mail: (DWW); (JY)
| | - Jiangtao Yan
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- * E-mail: (DWW); (JY)
| |
Collapse
|
53
|
Persson SE, Boman K, Wanhainen A, Carlberg B, Arnerlöv C. Decreasing prevalence of abdominal aortic aneurysm and changes in cardiovascular risk factors. J Vasc Surg 2017; 65:651-658. [DOI: 10.1016/j.jvs.2016.08.091] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 08/24/2016] [Indexed: 12/11/2022]
|
54
|
Shen YH, LeMaire SA. Molecular pathogenesis of genetic and sporadic aortic aneurysms and dissections. Curr Probl Surg 2017; 54:95-155. [PMID: 28521856 DOI: 10.1067/j.cpsurg.2017.01.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 01/16/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Ying H Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX.
| | - Scott A LeMaire
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX; Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX; Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX.
| |
Collapse
|
55
|
Liberale L, Dallegri F, Montecucco F, Carbone F. Pathophysiological relevance of macrophage subsets in atherogenesis. Thromb Haemost 2017; 117:7-18. [PMID: 27683760 DOI: 10.1160/th16-08-0593] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 09/12/2016] [Indexed: 12/14/2022]
Abstract
Macrophages are highly heterogeneous and plastic cells. They were shown to play a critical role in all stages of atherogenesis, from the initiation to the necrotic core formation and plaque rupture. Lesional macrophages primarily derive from blood monocyte, but local macrophage proliferation as well as differentiation from smooth muscle cells have also been described. Within atherosclerotic plaques, macrophages rapidly respond to changes in the microenvironment, shifting between pro- (M1) or anti-inflammatory (M2) functional phenotypes. Furthermore, different stimuli have been associated with differentiation of newly discovered M2 subtypes: IL-4/IL-13 (M2a), immune-complex (M2b), IL-10/glucocorticoids (M2c), and adenosine receptor agonist (M2d). More recently, additional intraplaque macrophage phenotypes were also recognized in response to CXCL4 (M4), oxidized phospholipids (Mox), haemoglobin/haptoglobin complexes (HA-mac/M(Hb)), and heme (Mhem). Such macrophage polarization was described as a progression among multiple phenotypes, which reflect the activity of different transcriptional factors and the cross-talk between intracellular signalling. Finally, the distribution of macrophage subsets within different plaque areas was markedly associated with cardiovascular (CV) vulnerability. The aim of this review is to update the current knowledge on the role of macrophage subsets in atherogenesis. In addition, the molecular mechanisms underlying macrophage phenotypic shift will be summarised and discussed. Finally, the role of intraplaque macrophages as predictors of CV events and the therapeutic potential of these cells will be discussed.
Collapse
Affiliation(s)
| | | | - Fabrizio Montecucco
- Fabrizio Montecucco, MD, PhD, Department of Internal Medicine, University of Genoa School of Medicine, 6 viale Benedetto XV, 16132 Genoa, Italy, Tel.: +39 010 353 8694, Fax: +39 010 353 8686, E-mail:
| | | |
Collapse
|
56
|
Atorvastatin Improves Ventricular Remodeling after Myocardial Infarction by Interfering with Collagen Metabolism. PLoS One 2016; 11:e0166845. [PMID: 27880844 PMCID: PMC5120826 DOI: 10.1371/journal.pone.0166845] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 11/05/2016] [Indexed: 12/27/2022] Open
Abstract
Purpose Therapeutic strategies that modulate ventricular remodeling can be useful after acute myocardial infarction (MI). In particular, statins may exert effects on molecular pathways involved in collagen metabolism. The aim of this study was to determine whether treatment with atorvastatin for 4 weeks would lead to changes in collagen metabolism and ventricular remodeling in a rat model of MI. Methods Male Wistar rats were used in this study. MI was induced in rats by ligation of the left anterior descending coronary artery (LAD). Animals were randomized into three groups, according to treatment: sham surgery without LAD ligation (sham group, n = 14), LAD ligation followed by 10mg atorvastatin/kg/day for 4 weeks (atorvastatin group, n = 24), or LAD ligation followed by saline solution for 4 weeks (control group, n = 27). After 4 weeks, hemodynamic characteristics were obtained by a pressure-volume catheter. Hearts were removed, and the left ventricles were subjected to histologic analysis of the extents of fibrosis and collagen deposition, as well as the myocyte cross-sectional area. Expression levels of mediators involved in collagen metabolism and inflammation were also assessed. Results End-diastolic volume, fibrotic content, and myocyte cross-sectional area were significantly reduced in the atorvastatin compared to the control group. Atorvastatin modulated expression levels of proteins related to collagen metabolism, including MMP1, TIMP1, COL I, PCPE, and SPARC, in remote infarct regions. Atorvastatin had anti-inflammatory effects, as indicated by lower expression levels of TLR4, IL-1, and NF-kB p50. Conclusion Treatment with atorvastatin for 4 weeks was able to attenuate ventricular dysfunction, fibrosis, and left ventricular hypertrophy after MI in rats, perhaps in part through effects on collagen metabolism and inflammation. Atorvastatin may be useful for limiting ventricular remodeling after myocardial ischemic events.
Collapse
|
57
|
Yang N, Cheng W, Hu H, Xue M, Li X, Wang Y, Xuan Y, Li X, Yin J, Shi Y, Yan S. Atorvastatin attenuates sympathetic hyperinnervation together with the augmentation of M2 macrophages in rats postmyocardial infarction. Cardiovasc Ther 2016; 34:234-44. [PMID: 27149420 DOI: 10.1111/1755-5922.12193] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Affiliation(s)
- Na Yang
- Department of Cardiology; School of Medicine; Shandong University; Ji'nan Shandong China
| | - Wenjuan Cheng
- Department of Cardiology; Shandong Provincial Qianfoshan Hospital; Ji'nan Shandong China
| | - Hesheng Hu
- Department of Cardiology; Shandong Provincial Qianfoshan Hospital; Ji'nan Shandong China
| | - Mei Xue
- Department of Cardiology; Shandong Provincial Qianfoshan Hospital; Ji'nan Shandong China
| | - Xiaolu Li
- Department of Cardiology; Shandong Provincial Qianfoshan Hospital; Ji'nan Shandong China
| | - Ye Wang
- Department of Cardiology; Shandong Provincial Qianfoshan Hospital; Ji'nan Shandong China
| | - Yongli Xuan
- Department of Cardiology; School of Medicine; Shandong University; Ji'nan Shandong China
| | - Xinran Li
- Department of Cardiology; School of Medicine; Shandong University; Ji'nan Shandong China
| | - Jie Yin
- Department of Cardiology; School of Medicine; Shandong University; Ji'nan Shandong China
| | - Yugen Shi
- Department of Cardiology; School of Medicine; Shandong University; Ji'nan Shandong China
| | - Suhua Yan
- Department of Cardiology; Shandong Provincial Qianfoshan Hospital; Ji'nan Shandong China
| |
Collapse
|
58
|
Activation of the vitamin D receptor selectively interferes with calcineurin-mediated inflammation: a clinical evaluation in the abdominal aortic aneurysm. J Transl Med 2016; 96:784-90. [PMID: 27239732 DOI: 10.1038/labinvest.2016.55] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 03/17/2016] [Accepted: 03/29/2016] [Indexed: 12/23/2022] Open
Abstract
In vitro and in vivo studies attribute potent immune regulatory properties to the vitamin D receptor (VDR). Yet, it is unclear to what extend these observations translate to the clinical context of (vascular) inflammation. This clinical study evaluates the potential of a VDR agonist to quench vascular inflammation. Patients scheduled for open abdominal aneurysm repair received paricalcitol 1 μg daily during 2-4 weeks before repair. Results were compared with matched controls. Evaluation in a parallel group showed that AAA patients are vitamin D insufficient (median plasma vitamin D: 43 (30-62 (IQR)) nmol/l). Aneurysm wall samples were collected during surgery, and the inflammatory footprint was studied. The brief paricalcitol intervention resulted in a selective 73% reduction in CD4+ T-helper cell content (P<0.024) and a parallel 35% reduction in T-cell (CD3+) content (P<0.032). On the mRNA level, paricalcitol reduced expression of T-cell-associated cytokines IL-2, 4, and 10 (P<0.019). No effect was found on other inflammatory mediators. On the protease level, selective effects were found for cathepsin K (P<0.036) and L (P<0.005). Collectively, these effects converge at the level of calcineurin activity. An effect of the VDR agonist on calcineurin activity was confirmed in a mixed lymphocyte reaction. In conclusion, brief course of the VDR agonist paricalcitol has profound effects on local inflammation via reduced T-cell activation. The anti-inflammatory potential of VDR activation in vitamin D insufficient patients is highly selective and appears to be mediated by an effect on calcineurin-mediated responses.
Collapse
|
59
|
IL-6: A Janus-like factor in abdominal aortic aneurysm disease. Atherosclerosis 2016; 251:139-146. [PMID: 27318834 DOI: 10.1016/j.atherosclerosis.2016.06.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 06/08/2016] [Accepted: 06/09/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS An abdominal aortic aneurysm (AAA) is part of the atherosclerotic spectrum of diseases. The disease is hallmarked by a comprehensive localized inflammatory response with striking IL-6 hyperexpression. IL-6 is a multifaceted cytokine that, depending on the context, acts as a pro- or anti-inflammatory factor. In this study, we explore a putative role for IL-6 in AAA disease. METHODS ELISA's, Western blot analysis, real time PCR and array analysis were used to investigate IL-6 expression and signaling in aneurysm wall samples from patients undergoing elective AAA repair. A role for IL-6 in AAA disease was tested through IL-6 neutralization experiments (neutralizing antibody) in the elastase model of AAA disease. RESULTS We confirmed an extreme disparity in aortic wall IL-6 content between AAA and atherosclerotic disease (median [5th-95th percentile] aortic wall IL-6 content: 281.6 [0.0-1820.8] (AAA) vs. 1.9 [0.0-37.8] μg/g protein (atherosclerotic aorta), (p < 0.001). Array analysis followed by pathway analysis showed that IL-6 hyper-expression is followed by increased IL-6 signaling (p < 0.000039), an observation confirmed by higher aneurysm wall pSTAT3 levels, and SOCS1 and SOCS3 mRNA expression, (p < 0.018). Remarkably, preventive IL-6 neutralization i.e. treatment started one day prior to the elastase-induction resulted in >40% 7-day mortality due to aortic rupture. In contrast, delayed IL-6 neutralization (i.e. neutralization started at day 4 after elastase induction) did not result in ruptures, and quenched AAA growth (p < 0.021). CONCLUSIONS AAA disease is characterized by increased IL-6 signaling. In the context of the elastase model of AAA disease, IL-6 appears a multi-faceted factor, protective upon acute injury, but negatively involved in the perpetuation of the disease process.
Collapse
|
60
|
Priebe HJ. Pharmacological modification of the perioperative stress response in noncardiac surgery. Best Pract Res Clin Anaesthesiol 2016; 30:171-89. [DOI: 10.1016/j.bpa.2016.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 03/21/2016] [Accepted: 03/31/2016] [Indexed: 11/26/2022]
|
61
|
Salic K, Morrison MC, Verschuren L, Wielinga PY, Wu L, Kleemann R, Gjorstrup P, Kooistra T. Resolvin E1 attenuates atherosclerosis in absence of cholesterol-lowering effects and on top of atorvastatin. Atherosclerosis 2016; 250:158-65. [PMID: 27236706 DOI: 10.1016/j.atherosclerosis.2016.05.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 04/26/2016] [Accepted: 05/01/2016] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND AIMS Besides LDL-cholesterol, local vascular inflammation plays a key role in atherogenesis. Efficient therapies to treat the inflammatory component of the disease have not been established. The discovery of specialized inflammation-resolving mediators, such as resolvins may provide new opportunities for treatment. This study examines whether the ω-3 fatty acid eicosapentaenoic acid-derived resolvin E1 (RvE1), can reduce atherosclerosis, when administered alone or in combination with a cholesterol-lowering statin. METHODS ApoE*3Leiden mice were fed a hypercholesterolemic diet for 9 weeks and subsequently treated with RvE1-low (1 mg/kg/day), RvE1-high (5 mg/kg/day), atorvastatin (1.5 mg/kg/day) or the combination of atorvastatin and RvE1-low for the following 16 weeks. RESULTS RvE1-low and RvE1-high reduced atherosclerotic lesion size to the same extent (-35%; p < 0.05), attenuated the formation of severe lesions, also seen as a proportional increase in the presence of mild lesions, but did not alter plasma cholesterol levels. Cholesterol-lowering atorvastatin reduced atherosclerosis (-27%, p < 0.05), and the combination of RvE1 and atorvastatin further attenuated lesion size (-51%, p < 0.01) and increased the content of mild lesions. RvE1 did not affect plasma SAA, E-selectin, VCAM-1 or MCP-1 but did reduce plasma EPHX4 and down-regulated the local expression of pro-atherogenic genes in the aortae, (e.g. Cd74, Cd44, Ccl2, Ccr5 and Adam17) and significantly inactivated IFN-γ (p < 0.001) and TNF-α (p < 0.001) signalling pathways. CONCLUSIONS RvE1 attenuates atherogenesis both alone and on top of a statin. The local effects of RvE1 are demonstrated by the modulated aortic expression of genes involved in inflammatory and immune responses, without altering plasma cholesterol or circulating SAA.
Collapse
Affiliation(s)
- Kanita Salic
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Zernikedreef 9, 2333 CK, Leiden, The Netherlands.
| | - Martine C Morrison
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Zernikedreef 9, 2333 CK, Leiden, The Netherlands
| | - Lars Verschuren
- Department of Microbiology and Systems Biology, Netherlands Organisation for Applied Scientific Research (TNO), Utrechtseweg 48, 3704 HE, Zeist, The Netherlands
| | - Peter Y Wielinga
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Zernikedreef 9, 2333 CK, Leiden, The Netherlands
| | - Lijun Wu
- Resolvyx Pharmaceuticals, Inc., 222 Third Street, Cambridge, MA, 02142, United States
| | - Robert Kleemann
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Zernikedreef 9, 2333 CK, Leiden, The Netherlands
| | - Per Gjorstrup
- Resolvyx Pharmaceuticals, Inc., 222 Third Street, Cambridge, MA, 02142, United States.
| | - Teake Kooistra
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Zernikedreef 9, 2333 CK, Leiden, The Netherlands
| |
Collapse
|
62
|
Metformin treatment status and abdominal aortic aneurysm disease progression. J Vasc Surg 2016; 64:46-54.e8. [PMID: 27106243 DOI: 10.1016/j.jvs.2016.02.020] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 02/01/2016] [Indexed: 12/11/2022]
Abstract
OBJECTIVE In population-based studies performed on multiple continents during the past two decades, diabetes mellitus has been negatively associated with the prevalence and progression of abdominal aortic aneurysm (AAA) disease. We investigated the possibility that metformin, the primary oral hypoglycemic agent in use worldwide, may influence the progression of AAA disease. METHODS Preoperative AAA patients with diabetes were identified from an institutional database. After tabulation of individual cardiovascular and demographic risk factors and prescription drug regimens, odds ratios for categorical influences on annual AAA enlargement were calculated through nominal logistical regression. Experimental AAA modeling experiments were subsequently performed in normoglycemic mice to validate the database-derived observations as well as to suggest potential mechanisms of metformin-mediated aneurysm suppression. RESULTS Fifty-eight patients met criteria for study inclusion. Of 11 distinct classes of medication considered, only metformin use was negatively associated with AAA enlargement. This association remained significant after controlling for gender, age, cigarette smoking status, and obesity. The median enlargement rate in AAA patients not taking oral diabetic medication was 1.5 mm/y; by nominal logistic regression, metformin, hyperlipidemia, and age ≥70 years were associated with below-median enlargement, whereas sulfonylurea therapy, initial aortic diameter ≥40 mm, and statin use were associated with above-median enlargement. In experimental modeling, metformin dramatically suppressed the formation and progression, with medial elastin and smooth muscle preservation and reduced aortic mural macrophage, CD8 T cell, and neovessel density. CONCLUSIONS Epidemiologic evidence of AAA suppression in diabetes may be attributable to concurrent therapy with the oral hypoglycemic agent metformin.
Collapse
|
63
|
Nenna A, Spadaccio C, Prestipino F, Lusini M, Sutherland FW, Beattie GW, Petitti T, Nappi F, Chello M. Effect of Preoperative Aspirin Replacement With Enoxaparin in Patients Undergoing Primary Isolated On-Pump Coronary Artery Bypass Grafting. Am J Cardiol 2016; 117:563-570. [PMID: 26721653 DOI: 10.1016/j.amjcard.2015.11.040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/19/2015] [Accepted: 11/19/2015] [Indexed: 10/22/2022]
Abstract
Management of preoperative antiplatelet therapy in coronary artery bypass grafting (CABG) is variable among surgeons: guidelines collide with prejudices because replacement of aspirin with low-molecular-weight heparin is still performed because of a presumed minor bleeding risk. This study aims to analyze postoperative bleedings and complications in patients scheduled for elective primary isolated on-pump CABG, depending on preoperative aspirin treatment or its replacement with enoxaparin. In this cohort study, we propensity score matched 200 patients in whom aspirin was stopped at least 5 days before CABG and replaced with enoxaparin and 200 patients who continued aspirin therapy until the day before surgery. Postoperative bleedings and complications were monitored during hospitalization. Among patients who continued aspirin treatment, mean overall bleeding was 701.0 ± 334.6 ml, whereas in the matched enoxaparin group, it was significantly greater (882.6 ± 64.6 ml, p value <0.001); this was associated with reduced postoperative complications, lower values of postoperative C-reactive protein in aspirin takers, and a presumed protective effect for statins. After propensity score adjustment, aspirin treatment carried a protective effect against major postoperative bleeding (odds ratio 0.312, p = 0.001). In conclusion, postoperative bleeding is reduced in patients who continued aspirin, likely due to a reduction in postoperative inflammation. The practice of empirically discontinuing aspirin and replacing it with enoxaparin before CABG should be abandoned. Patients with coronary artery disease referred to CABG should continue antiplatelet medications until the surgical procedure. Those results might be extended to patients under oral anticoagulant therapy requiring CABG.
Collapse
|
64
|
Garip S, Bayari SH, Severcan M, Abbas S, Lednev IK, Severcan F. Structural effects of simvastatin on liver rat [corrected] tissue: Fourier transform infrared and Raman microspectroscopic studies. JOURNAL OF BIOMEDICAL OPTICS 2016; 21:25008. [PMID: 26891599 DOI: 10.1117/1.jbo.21.2.025008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 01/21/2016] [Indexed: 06/05/2023]
Abstract
Simvastatin is one of the most frequently prescribed statins because of its efficacy in the treatment of hypercholesterolemia, reducing cardiovascular risk and related mortality. Determination of its side effects on different tissues is mandatory to improve safe use of this drug. In the present study, the effects of simvastatin on molecular composition and structure of healthy rat livers were investigated by Fourier transform infrared and Raman imaging. Simvastatin-treated groups received 50 mg/kg/day simvastatin for 30 days. The ratio of the area and/or intensity of the bands assigned to lipids, proteins, and nucleic acids were calculated to get information about the drug-induced changes in tissues. Loss of unsaturation, accumulation of end products of lipid peroxidation, and alterations in lipid-to-protein ratio were observed in the treated group. Protein secondary structure studies revealed significant decrease in α-helix and increase in random coil, while native β-sheet decreases and aggregated β-sheet increases in treated group implying simvastatin-induced protein denaturation. Moreover, groups were successfully discriminated using principal component analysis. Consequently, high-dose simvastatin treatment induces hepatic lipid peroxidation and changes in molecular content and protein secondary structure, implying the risk of liver disorders in drug therapy.
Collapse
Affiliation(s)
- Sebnem Garip
- Istanbul Kemerburgaz University, Department of Medical Biochemistry, Faculty of Medicine, Mahmutbey Dilmenler Caddesi, No: 26, Istanbul 34217, Turkey
| | - Sevgi Haman Bayari
- Hacettepe University, Department of Physics Engineering, Faculty of Engineering, Beytepe Campus, Ankara 06800, Turkey
| | - Mete Severcan
- Middle East Technical University, Department of Electrical and Electronics Engineering, Faculty of Engineering, Dumlupinar Bulvari, No: 1, Ankara 06800, Turkey
| | - Sherif Abbas
- Middle East Technical University, Department of Biological Sciences, Faculty of Arts and Sciences, Dumlupinar Bulvari, No: 1, Ankara 06800, Turkey
| | - Igor K Lednev
- University at Albany, Department of Chemistry, State University of New York, Albany, New York 12222, United States
| | - Feride Severcan
- University at Albany, Department of Chemistry, State University of New York, Albany, New York 12222, United States
| |
Collapse
|
65
|
Deeg MA, Meijer CA, Chan LS, Shen L, Lindeman JHN. Prognostic and predictive biomarkers of abdominal aortic aneurysm growth rate. Curr Med Res Opin 2016; 32:509-17. [PMID: 26636178 DOI: 10.1185/03007995.2015.1128406] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
OBJECTIVES To test the utility of clinical and circulating biomarkers to predict abdominal aortic aneurysm (AAA) growth rate and response to doxycycline therapy. METHODS Plasma samples were obtained in the Pharmaceutical Aneurysm Stabilization Trial that tested the effect of doxycycline (n = 44) vs. placebo (n = 49) in patients with a 35-50 mm AAA. Approximately 200 biomarkers were evaluated in a candidate approach that included markers of matrix turnover and cathepsin S activity and a broad-based approach of predominantly inflammation-related and clinical biomarkers. RESULTS In a recursive partitioning based analysis, total cholesterol, baseline AAA size, and apolipoprotein B were prognostic of AAA growth in the placebo group whereas elastin and biglycan degradation products were predictive of AAA growth with doxycycline treatment. Univariate analysis of these biomarkers showed that baseline total cholesterol (r = 0.38, unadjusted P = 0.011), apolipoprotein B (r = 0.41, unadjusted P = 0.005), and baseline AAA size (r = 0.35, unadjusted P = 0.013) correlated with AAA growth in the placebo but not the doxycycline group. Elastin fragments were associated with 18 month AAA growth (r = 0.33, unadjusted P = 0.031) in the doxycycline group. LIMITATIONS Limitations of this study include small sample size, a retrospective growth analysis, and translatability of the method used to measure the analytes. CONCLUSIONS This study implies that total cholesterol, baseline AAA size, and apolipoprotein B are predictors of AAA growth. Levels of elastin and biglycan fragments are predictive of doxycycline effects on AAA growth and provide a clue towards this unexpected negative effect.
Collapse
Affiliation(s)
- Mark A Deeg
- a a Lilly Research Laboratories, Eli Lilly & Co. , Indianapolis , IN , USA
| | - C Arnoud Meijer
- b b Department of Vascular Surgery , Leiden University Medical Center , Leiden , The Netherlands
| | - Lai Shan Chan
- a a Lilly Research Laboratories, Eli Lilly & Co. , Indianapolis , IN , USA
| | - Lei Shen
- a a Lilly Research Laboratories, Eli Lilly & Co. , Indianapolis , IN , USA
| | - Jan H N Lindeman
- b b Department of Vascular Surgery , Leiden University Medical Center , Leiden , The Netherlands
| |
Collapse
|
66
|
Kokje VBC, Hamming JF, Lindeman JHN. Editor's Choice - Pharmaceutical Management of Small Abdominal Aortic Aneurysms: A Systematic Review of the Clinical Evidence. Eur J Vasc Endovasc Surg 2015; 50:702-13. [PMID: 26482507 DOI: 10.1016/j.ejvs.2015.08.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 08/15/2015] [Indexed: 11/27/2022]
Abstract
BACKGROUND Management of abdominal aortic aneurysms (AAAs) relies on surgical repair of larger AAAs. Consequently medical interventions inhibiting AAA progression could greatly reduce the need for surgical repair. A spectrum of pharmaceutical strategies has been reported, albeit conclusions often appear contradictory. Given the longstanding interest in pharmaceutical AAA stabilization, a systematic review of the available literature is relevant. OBJECTIVES The aim is to provide an up to date systematic review of the available data on pharmaceutical therapies for stabilizing or impeding AAA growth. METHODS A search using Pubmed, Embase, Web of science, Cochrane, CINAHL, Academic Search Premier, and Science Direct identified 27 eligible papers that studied the clinical effect of the pharmaceutical therapy on AAA diameter growth. RESULTS This review shows that there is currently no pharmaceutical strategy that reduces AAA growth. Most studies are of poor methodological quality. Initial promising reports are often not confirmed in subsequent larger studies, raising the possibility of selective reporting. CONCLUSION There is currently no pharmaceutical means that halts AAA growth.
Collapse
Affiliation(s)
- V B C Kokje
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - J F Hamming
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - J H N Lindeman
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
67
|
Zhang W, Liu Z, Liu C. Effect of lipid-modifying therapy on long-term mortality after abdominal aortic aneurysm repair: a systemic review and meta-analysis. World J Surg 2015; 39:794-801. [PMID: 25385163 DOI: 10.1007/s00268-014-2858-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Several observational studies have demonstrated that lipid-modifying therapy may improve long-term survival in abdominal aortic aneurysm (AAA) patients after repair. We carried out a systematic review and meta-analysis of studies evaluating the effect of lipid-modifying therapy on long-term mortality. PATIENTS AND METHODS We conducted a systematic search of multiple databases up to April 2014. Studies that evaluated exposure to lipid-modifying therapy, reported mortality data and hazard ratio (HR) or provided survival curve for their estimation were included in the meta-analysis. Pooled HR estimates with 95% confidence intervals (CIs) were calculated using the random-effects model. RESULTS Eight studies (seven cohorts, one post hoc study of a randomization controlled trial) reporting 2,605 patients on lipid-modifying therapy were included. Meta-analysis showed a significant 39% reduction in long-term mortality with lipid-modifying therapy (HR 0.61; 95% CI 0.51-0.73). After exclusion of one study which was contributing to considerable heterogeneity, a significant 33% reduction in mortality risk was a more conservative, consistent estimate (HR 0.67; 95% CI 0.59-0.77). CONCLUSION Meta-analysis of studies supports a protective role of lipid-modifying therapy on mortality risk after AAA repair. Aggressive lipid intervention should be recommended to those who receiving AAA repair.
Collapse
Affiliation(s)
- Wenwen Zhang
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, People's Republic of China
| | | | | |
Collapse
|
68
|
Yu HH, Chen PC, Yang YH, Wang LC, Lee JH, Lin YT, Chiang BL. Statin reduces mortality and morbidity in systemic lupus erythematosus patients with hyperlipidemia: A nationwide population-based cohort study. Atherosclerosis 2015; 243:11-8. [PMID: 26342937 DOI: 10.1016/j.atherosclerosis.2015.08.030] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 08/10/2015] [Accepted: 08/22/2015] [Indexed: 12/28/2022]
Abstract
OBJECTIVE The anti-inflammatory and cardiovascular protective effects of statin for patients with systemic lupus erythematosus (SLE) are not clear. We tested the hypothesis that statin use is associated with reduced mortality and morbidity in SLE patients with hyperlipidemia. METHODS We included 4095 patients with SLE and hyperlipidemia from the entire population using the Taiwan National Health Insurance Research Database between 1997 and 2008. A total of 935 matching sets (1:2) of patients who had never used lipid-lowering medications and statin users were included in the nested matched cohort. Cox proportional hazards regression was used to calculate the hazard ratios (HR) and 95% confidence intervals (CI) for the association between statin and all-cause mortality, coronary artery disease (CAD), cerebrovascular disease (CVD) and end-stage renal disease (ESRD), conditional for matching sets in the matched cohort. RESULTS The multivariate adjusted hazard ratios (HR) for statin users, as compared with patients had never used lipid-lowering medications, were 0.67 (95% CI, 0.54 to 0.83) for death from any cause. High-dose statins (>365 cumulative defined daily dose) significantly reduced risk of all-cause mortality (HR 0.44, 95% CI 0.32 to 0.60); CAD (HR 0.20, 95% CI 0.13 to 0.31); CVD (HR 0.14, 95% CI 0.08 to 0.25); and ESRD (HR 0.22, 95% CI, 0.16 to 0.29), with similar results in the nested matched study. CONCLUSION Statin therapy in SLE patients with hyperlipidemia may reduce the risk of mortality, cardiovascular disease and ESRD. The effect of statins needs to be demonstrated in large prospective studies with long-term follow-up.
Collapse
Affiliation(s)
- Hsin-Hui Yu
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Pau-Chung Chen
- Institute of Occupational Medicine and Industrial Hygiene, National Taiwan University College of Public Health, Taipei, Taiwan
| | - Yao-Hsu Yang
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Li-Chieh Wang
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Jyh-Hong Lee
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yu-Tsan Lin
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Bor-Luen Chiang
- Department of Medical Research, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
69
|
Wang L, Peng J, Huang H, Wang Q, Yu M, Tao L. Simvastatin protects Sertoli cells against cisplatin cytotoxicity through enhanced gap junction intercellular communication. Oncol Rep 2015; 34:2133-41. [PMID: 26260290 DOI: 10.3892/or.2015.4192] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 07/09/2015] [Indexed: 11/06/2022] Open
Abstract
Cisplatin, an important chemotherapeutic agent against testicular germ cell cancer, induces testicular toxicity on Leydig and Sertoli cells, leading to serious side-effects such as azoospermia and infertility. In a previous study, it was found that simvastatin enhanced the sensitivity of Leydig tumor cells to chemotherapeutic toxicity through the enhancement of gap junction functions. In the present study, the effect of simvastatin on the sensitivity of normal Sertoli cells to cisplatin and the role of gap junctions in such effects was investigated. The results showed that, simvastatin attenuated cisplatin toxicity only when cells exhibited high-density culture where gap junctional formation was possible. When gap junction function was decreased by the gap junction inhibitor or by siRNA targeting connexin 43, the protective effect of simvastatin to cisplatin toxicity was substantially attenuated. Simvastatin also enhanced gap junction functions between Sertoli cells. This effect was mediated by the reduction of PKC-mediated connexin phosphorylation, thereby increasing connexin 43 membrane localization. Thus, simvastatin-induced enhancement of gap junction‑mediated intercellular communication attenuated cisplatin toxicity on Sertoli cells. This result indicated that enhancement of gap junction function by simvastatin may have bilateral beneficial effects on cisplatin‑based chemotherapy, enhancing cisplatin killing on cancer while ameliorating the reproduction toxicity.
Collapse
Affiliation(s)
- Lingzhi Wang
- Department of Anaesthesia, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, P.R. China
| | - Jianxin Peng
- Department of Hepatobiliary Surgery, Guangdong Province Traditional Chinese Medical Hospital, Guangzhou 510120, P.R. China
| | - Huansen Huang
- Department of Anaesthesia, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, P.R. China
| | - Qin Wang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, P.R. China
| | - Meiling Yu
- Department of Pharmacy, The First Affiliated Hospital, Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Liang Tao
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, P.R. China
| |
Collapse
|
70
|
|
71
|
Janssen H, Wagner CS, Demmer P, Callies S, Sölter G, Loghmani-khouzani H, Hu N, Schuett H, Tietge UJF, Warnecke G, Larmann J, Theilmeier G. Acute perioperative-stress-induced increase of atherosclerotic plaque volume and vulnerability to rupture in apolipoprotein-E-deficient mice is amenable to statin treatment and IL-6 inhibition. Dis Model Mech 2015; 8:1071-80. [PMID: 26092124 PMCID: PMC4582096 DOI: 10.1242/dmm.018713] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 06/09/2015] [Indexed: 01/15/2023] Open
Abstract
Myocardial infarction and stroke are frequent after surgical procedures and consume a considerable amount of benefit of surgical therapy. Perioperative stress, induced by surgery, is composed of hemodynamic and inflammatory reactions. The effects of perioperative stress on atherosclerotic plaques are ill-defined. Murine models to investigate the influence of perioperative stress on plaque stability and rupture are not available. We developed a model to investigate the influence of perioperative stress on plaque growth and stability by exposing apolipoprotein-E-deficient mice, fed a high cholesterol diet for 7 weeks, to a double hit consisting of 30 min of laparotomy combined with a substantial blood loss (approximately 20% of total blood volume; 400 µl). The innominate artery was harvested 72 h after the intervention. Control groups were sham and baseline controls. Interleukin-6 (IL-6) and serum amyloid A (SAA) plasma levels were determined. Plaque load, vascular smooth muscle cell (VSMC) and macrophage content were quantified. Plaque stability was assessed using the Stary score and frequency of signs of plaque rupture were assessed. High-dose atorvastatin (80 mg/kg body weight/day) was administered for 6 days starting 3 days prior to the double hit. A single dose of an IL-6-neutralizing antibody or the fusion protein gp130-Fc selectively targeting IL-6 trans-signaling was subcutaneously injected. IL-6 plasma levels increased, peaking at 6 h after the intervention. SAA levels peaked at 24 h (n=4, P<0.01). Plaque volume increased significantly with the double hit compared to sham (n=8, P<0.01). More plaques were scored as complex or bearing signs of rupture after the double hit compared to sham (n=5-8, P<0.05). Relative VSMC and macrophage content remained unchanged. IL-6-inhibition or atorvastatin, but not blocking of IL-6 trans-signaling, significantly decreased plaque volume and complexity (n=8, P<0.01). Using this model, researchers will be able to further investigate the pathophysiology of perioperative plaque stability, which can result in myocardial infarction, and, additionally, to test potential protective strategies. Summary: We developed a model to study the dynamics of atherosclerotic plaque growth and stability following surgery, and show that IL-6 inhibition and statins beneficially affect plaque volume and complexity.
Collapse
Affiliation(s)
- Henrike Janssen
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany Department of Anesthesiology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Christian S Wagner
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Philipp Demmer
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Simone Callies
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Gesine Sölter
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Houra Loghmani-khouzani
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Niandan Hu
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Harald Schuett
- Department of Cardiology, University Hospital Marburg, 35043 Marburg, Germany
| | - Uwe J F Tietge
- Department of Pediatrics, University of Groningen, UMCG, NL-9700 Groningen, The Netherlands
| | - Gregor Warnecke
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany German Centre for Lung Research (DZL), 30625 Hannover, Germany
| | - Jan Larmann
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany Department of Anesthesiology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Gregor Theilmeier
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany Faculty VI - Medicine and Health Sciences, Dept of Health Services Sciences, University of Oldenburg, 26129 Germany
| |
Collapse
|
72
|
Flessenkaemper IH, Loddenkemper R, Roll S, Enke-Melzer K, Wurps H, Bauer TT. Screening of COPD patients for abdominal aortic aneurysm. Int J Chron Obstruct Pulmon Dis 2015; 10:1085-91. [PMID: 26089658 PMCID: PMC4468935 DOI: 10.2147/copd.s81439] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Purpose Screening for abdominal aortic aneurysm (AAA) in “men aged over 65 years who have ever smoked” is a recommended policy. To reduce the number of screenings, it may be of value to define subgroups with a higher prevalence of AAA. Since chronic obstructive pulmonary disease (COPD) and AAA are associated with several common risk factors, this study investigates the prevalence of AAA in COPD patients. Patients and methods Patients with COPD were identified via the hospital information system. Inclusion criteria were: COPD stage I–IV, ability to give full consent, and age >18 years; exclusion criteria were: patient too obese for an ultrasound check, previously diagnosed AAA, prior surgery for AAA, or ethical grounds such as concomitant advanced malignant or end-stage disease. The primary endpoint of the study was an aortic diameter measured by ultrasound of ≥30 mm. Defined secondary endpoints were evaluated on the basis of medical records and interviews. Results Of the 1,180 identified COPD patients, 589 were included in this prospective study. In 22 patients (3.70%), the aortic diameter was ≥30 mm, representing an AAA prevalence of 6.72% among males aged >65 years. The risk of AAA increased with the following comorbidities/risk factors: male sex (odds ratio [OR] 2.98), coronary heart disease (OR 2.81), peripheral arterial occlusive disease (OR 2.47), hyperlipoproteinemia (OR 2.77), AAA in the family history (OR 3.95), and COPD stage I/II versus IV (OR 1.81). Conclusion The overall AAA prevalence of 3.7% in our group of COPD patients is similar to that of the general population aged >65 years. However, the frequency of AAA in male COPD patients aged >65 years is considerably higher (6.72%) and increased further still in those individuals with additional comorbidities/risk factors. Defining subgroups with a higher risk of AAA may increase the efficiency of screening.
Collapse
Affiliation(s)
| | | | - Stephanie Roll
- Institute for Social Medicine, Epidemiology and Health Economics, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Kathrin Enke-Melzer
- Department for Vascular Medicine, Helios Klinikum Emil von Behring, Berlin, Germany
| | - Henrik Wurps
- Department of Pneumology, Helios Klinikum Emil von Behring, Berlin, Germany
| | - Torsten T Bauer
- Department of Pneumology, Helios Klinikum Emil von Behring, Berlin, Germany
| |
Collapse
|
73
|
Lindeman JHN. The pathophysiologic basis of abdominal aortic aneurysm progression: a critical appraisal. Expert Rev Cardiovasc Ther 2015; 13:839-51. [PMID: 26028299 DOI: 10.1586/14779072.2015.1052408] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
An aneurysm of the abdominal aorta is a common pathology and a major cause of sudden death in the elderly. Currently, abdominal aortic aneurysms (AAAs) can only be treated by surgery and an effective medical therapy is urgently missing. The pathophysiology of AAAs is complex and is believed to be best described as a comprehensive inflammatory response with an accompanying proteolytic imbalance; the latter being held responsible for the progressive weakening of the aortic wall. Remarkably, while interference in inflammatory and/or proteolytic cascades proves highly effective in preclinical studies, emerging clinical studies consistently fail to show a benefit. In fact, some anti-inflammatory interventions appear to adversely influence the disease process. Altogether, recent clinical observations not only challenge the prevailing concepts of AAA progression, but also raise doubt on the translatability of findings from rodent models for growing AAA.
Collapse
Affiliation(s)
- Jan H N Lindeman
- Department Vascular and Transplant Surgery, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| |
Collapse
|
74
|
Piechota-Polanczyk A, Jozkowicz A, Nowak W, Eilenberg W, Neumayer C, Malinski T, Huk I, Brostjan C. The Abdominal Aortic Aneurysm and Intraluminal Thrombus: Current Concepts of Development and Treatment. Front Cardiovasc Med 2015; 2:19. [PMID: 26664891 PMCID: PMC4671358 DOI: 10.3389/fcvm.2015.00019] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 04/10/2015] [Indexed: 01/09/2023] Open
Abstract
The pathogenesis of the abdominal aortic aneurysm (AAA) shows several hallmarks of atherosclerotic and atherothrombotic disease, but comprises an additional, predominant feature of proteolysis resulting in the degradation and destabilization of the aortic wall. This review aims to summarize the current knowledge on AAA development, involving the accumulation of neutrophils in the intraluminal thrombus and their central role in creating an oxidative and proteolytic environment. Particular focus is placed on the controversial role of heme oxygenase 1/carbon monoxide and nitric oxide synthase/peroxynitrite, which may exert both protective and damaging effects in the development of the aneurysm. Treatment indications as well as surgical and pharmacological options for AAA therapy are discussed in light of recent reports.
Collapse
Affiliation(s)
- Aleksandra Piechota-Polanczyk
- Department of Surgery, Medical University of Vienna , Vienna , Austria ; Department of Biochemistry, Medical University of Lodz , Lodz , Poland
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Jagiellonian University , Krakow , Poland
| | - Witold Nowak
- Department of Medical Biotechnology, Jagiellonian University , Krakow , Poland
| | - Wolf Eilenberg
- Department of Surgery, Medical University of Vienna , Vienna , Austria
| | | | - Tadeusz Malinski
- Department of Chemistry and Biochemistry, Ohio University , Athens, OH , USA
| | - Ihor Huk
- Department of Surgery, Medical University of Vienna , Vienna , Austria
| | | |
Collapse
|
75
|
Kroon AM, Taanman JW. Clonal expansion of T cells in abdominal aortic aneurysm: a role for doxycycline as drug of choice? Int J Mol Sci 2015; 16:11178-95. [PMID: 25993290 PMCID: PMC4463695 DOI: 10.3390/ijms160511178] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 02/05/2015] [Indexed: 11/16/2022] Open
Abstract
Most reported studies with animal models of abdominal aortic aneurysm (AAA) and several studies with patients have suggested that doxycycline favourably modifies AAA; however, a recent large long-term clinical trial found that doxycycline did not limit aneurysm growth. Thus, there is currently no convincing evidence that doxycycline reduces AAA expansion. Here, we critically review the available experimental and clinical information about the effects of doxycycline when used as a pharmacological treatment for AAA. The view that AAA can be considered an autoimmune disease and the observation that AAA tissue shows clonal expansion of T cells is placed in the light of the well-known inhibition of mitochondrial protein synthesis by doxycycline. In T cell leukaemia animal models, this inhibitory effect of the antibiotic has been shown to impede T cell proliferation, resulting in complete tumour eradication. We suggest that the available evidence of doxycycline action on AAA is erroneously ascribed to its inhibition of matrix metalloproteinases (MMPs) by competitive binding of the zinc ion co-factor. Although competitive binding may explain the inhibition of proteolytic activity, it does not explain the observed decreases of MMP mRNA levels. We propose that the observed effects of doxycycline are secondary to inhibition of mitochondrial protein synthesis. Provided that serum doxycycline levels are kept at adequate levels, the inhibition will result in a proliferation arrest, especially of clonally expanding T cells. This, in turn, leads to the decrease of proinflammatory cytokines that are normally generated by these cells. The drastic change in cell type composition may explain the changes in MMP mRNA and protein levels in the tissue samples.
Collapse
Affiliation(s)
- Albert M Kroon
- Department of Clinical Neurosciences, Institute of Neurology, University College London, London NW3 2PF, UK.
| | - Jan-Willem Taanman
- Department of Clinical Neurosciences, Institute of Neurology, University College London, London NW3 2PF, UK.
| |
Collapse
|
76
|
Yoshimura K, Nagasawa A, Kudo J, Onoda M, Morikage N, Furutani A, Aoki H, Hamano K. Inhibitory effect of statins on inflammation-related pathways in human abdominal aortic aneurysm tissue. Int J Mol Sci 2015; 16:11213-28. [PMID: 25993292 PMCID: PMC4463697 DOI: 10.3390/ijms160511213] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 03/27/2015] [Accepted: 03/30/2015] [Indexed: 12/03/2022] Open
Abstract
HMG-CoA (3-hydroxy-3-methylglutaryl-coenzyme A) reductase inhibitors (statins) have been suggested to attenuate abdominal aortic aneurysm (AAA) growth. However, the effects of statins in human AAA tissues are not fully elucidated. The aim of this study was to investigate the direct effects of statins on proinflammatory molecules in human AAA walls in ex vivo culture. Simvastatin strongly inhibited the activation of nuclear factor (NF)-κB induced by tumor necrosis factor (TNF)-α in human AAA walls, but showed little effect on c-jun N-terminal kinase (JNK) activation. Simvastatin, as well as pitavastatin significantly reduced the secretion of matrix metalloproteinase (MMP)-9, monocyte chemoattractant protein (MCP)-2 and epithelial neutrophil-activating peptide (CXCL5) under both basal and TNF-α-stimulated conditions. Similar to statins, the Rac1 inhibitor NSC23766 significantly inhibited the activation of NF-κB, accompanied by a decreased secretion of MMP-9, MCP-2 and CXCL5. Moreover, the effect of simvastatin and the JNK inhibitor SP600125 was additive in inhibiting the secretion of MMP-9, MCP-2 and CXCL5. These findings indicate that statins preferentially inhibit the Rac1/NF-κB pathway to suppress MMP-9 and chemokine secretion in human AAA, suggesting a mechanism for the potential effect of statins in attenuating AAA progression.
Collapse
Affiliation(s)
- Koichi Yoshimura
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan.
- Graduate School of Health and Welfare, Yamaguchi Prefectural University, Yamaguchi 753-8502, Japan.
| | - Ayako Nagasawa
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan.
- Division of Thoracic and Cardiovascular Surgery, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8510, Japan.
| | - Junichi Kudo
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan.
| | - Masahiko Onoda
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan.
| | - Noriyasu Morikage
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan.
| | - Akira Furutani
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan.
| | - Hiroki Aoki
- Cardiovascular Research Institute, Kurume University, Kurume 830-0011, Japan.
| | - Kimikazu Hamano
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan.
| |
Collapse
|
77
|
Shen Q, Riedl KM, Cole RM, Lehman C, Xu L, Alder H, Belury MA, Schwartz SJ, Ziouzenkova O. Egg yolks inhibit activation of NF-κB and expression of its target genes in adipocytes after partial delipidation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:2013-25. [PMID: 25620076 PMCID: PMC4362627 DOI: 10.1021/jf5056584] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
How composition of egg yolk (EY) influences NF-κB, a key transcription pathway in inflammation, remains unclear. We performed partial delipidation of EY that removed 20-30% of cholesterol and triglycerides. The resulting polar and nonpolar fractions were termed EY-P and EY-NP. NF-κB activation in response to EY from different suppliers and their fractions was examined in 3T3-L1 adipocytes using a NF-κB response element reporter assay and by analyzing expression of 248 inflammatory genes. Although EY-P and EY contained similar level of vitamins, carotenoids, and fatty acids, only delipidated EY-P fraction suppressed NF-κB via down-regulation of toll like receptor-2 and up-regulation of inhibitory toll interacting protein (Tollip) and lymphocyte antigen 96 (Ly96). Our data suggest that anti-inflammatory activity of lutein and retinol were blunted by nonpolar lipids in EY, likely via crosstalk between SREBP and NF-κB pathways in adipocytes. Thus, moderate delipidation may improve the beneficial properties of regular eggs.
Collapse
Affiliation(s)
- Qiwen Shen
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Ken M. Riedl
- Department of Food Science and Technology, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Rachel M. Cole
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Christopher Lehman
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Lu Xu
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, 43210, USA
- Division of Minimally Invasive Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hansjuerg Alder
- Nucleic Acid Shared Resource, Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Martha A. Belury
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Steven J. Schwartz
- Department of Food Science and Technology, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Ouliana Ziouzenkova
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, 43210, USA
- Corresponding author: O.Z., 1787 Neil Avenue, 331A Campbell Hall, Columbus, OH, 43210, , Telephone: 614 292 5034, Fax: 614 292 8880
| |
Collapse
|
78
|
Carbone F, Montecucco F. Inflammation in arterial diseases. IUBMB Life 2015; 67:18-28. [PMID: 25631520 DOI: 10.1002/iub.1344] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 12/28/2014] [Indexed: 12/26/2022]
Abstract
The pathophysiology of some inflammatory arterial diseases (such as vasculitis, atherogenesis and aneurysms) has been widely investigated in the last decades. Among different soluble molecules, proinflammatory cytokines (such as TNF-α, IL-1 and IL-6) were shown to trigger critical pathways regulating these arterial diseases. Together with these cytokines, chemokines were also associated with endothelial dysfunction and intima injury in arterial diseases. Recently, autoantibodies have been also described to pathophysiologically influence not only autoimmune vasculitis but also atherogenesis and more in general vascular inflammation. These soluble mediators actively trigger inflammatory functions of leukocytes and vascular cells. For instance, B and T lymphocytes, macrophages and neutrophils were shown to actively participate in inflammatory processes within the arterial wall in vasculitis, atherogenesis and aneurysms. The aim of this narrative review is to provide an overview of pathophysiology and treatments targeting arterial inflammation in these diseases.
Collapse
Affiliation(s)
- Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa School of Medicine, IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy; Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, Geneva, Switzerland
| | | |
Collapse
|
79
|
Tedesco S, Bolego C, Toniolo A, Nassi A, Fadini GP, Locati M, Cignarella A. Phenotypic activation and pharmacological outcomes of spontaneously differentiated human monocyte-derived macrophages. Immunobiology 2014; 220:545-54. [PMID: 25582402 DOI: 10.1016/j.imbio.2014.12.008] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 12/03/2014] [Accepted: 12/15/2014] [Indexed: 01/16/2023]
Abstract
Macrophage activation has been observed in vivo under physiological and pathological conditions, and may represent an attractive target for pharmacological modulation. This study tested the hypothesis that human blood-derived macrophages generated in vitro in the absence of specific macrophage growth factors respond flexibly to activation stimuli and pharmacological treatment. Monocytes were differentiated to macrophages for 7 days in culture in RPMI 1640 with 10% FCS. The resulting population showed predominance of the M2 over M1 phenotype as measured by flow cytometry and the expression of M1 vs. M2 markers was not mutually exclusive. Activation with LPS/IFN-γ for 48 h significantly increased the fraction of surface CD68-expressing cells, the CD14(+)/CD16(-)/CD68(+) subset and cell-bound TNF-α levels, whereas expression of the CC chemokine receptor (CCR)-2 was unchanged. Expression of the M2 markers CD206, CD163 and CX3CR1 was down-regulated following M1 activation compared with resting and after pre-exposure to M2-triggers. By contrast, alternative activation with IL-4/IL-13 for 48 h did not increase M2 markers, while CD206 up-regulation was observed after 7 days. Both activation signals induced changes in gene expression profiles as shown by Q-PCR. Treatment with 100 nM dexamethasone enhanced the M2 morphotype and CD163 expression while preventing LPS/IFN-γ-induced CD163 down-regulation. After 1-week dexamethasone treatment, virtually all cells acquired a CD163(+)/CD206(+)/CX3CR1(+) M2 phenotype. Therefore, these protocols appear to be useful to perform screens of pharmacological agents targeting human macrophage activation.
Collapse
Affiliation(s)
- Serena Tedesco
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Italy
| | - Chiara Bolego
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Italy.
| | - Alice Toniolo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Italy
| | - Alberto Nassi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Italy
| | - Gian Paolo Fadini
- Department of Medicine, University Hospital, Italy; Venetian Institute of Molecular Medicine, Padua, Italy
| | - Massimo Locati
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Italy; Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Andrea Cignarella
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Italy
| |
Collapse
|
80
|
Kortekaas KE, Meijer CA, Hinnen JW, Dalman RL, Xu B, Hamming JF, Lindeman JH. ACE inhibitors potently reduce vascular inflammation, results of an open proof-of-concept study in the abdominal aortic aneurysm. PLoS One 2014; 9:e111952. [PMID: 25474105 PMCID: PMC4256371 DOI: 10.1371/journal.pone.0111952] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 09/29/2014] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Independent of their blood pressure lowering effect, ACE inhibitors are thought to reduce vascular inflammation. The clinical relevance of this effect is unclear with the current knowledge. Abdominal aortic aneurysms (AAA) are characterized by a broad, non-specific inflammatory response, and thus provide a clinical platform to evaluate the anti-inflammatory potential of ACE inhibitors. METHODS AND RESULTS Eleven patients scheduled for open AAA repair received ramipril (5 mg/day) during 2-4 weeks preceding surgery. Aortic wall samples were collected during surgery, and compared to matched samples obtained from a biobank. An anti-inflammatory potential was evaluated in a comprehensive analysis that included immunohistochemistry, mRNA and protein analysis. A putative effect of ACE inhibitors on AAA growth was tested separately by comparing 18-month growth rate of patients on ACE inhibitors (n = 82) and those not taking ACE inhibitors (n = 204). Ramipril reduces mRNA expression of multiple pro-inflammatory cytokines such as IL-1β, IL-6, IL-8, TNF -α, Interferon-[Formula: see text], and MCP-1, as well as aortic wall IL-8 and MCP-1 (P = 0.017 and 0.008, respectively) protein content. The is followed by clear effects on cell activation that included a shift towards anti-inflammatory macrophage (M2) subtype. Evaluation of data from the PHAST cohort did not indicate an effect of ACE inhibitors on 18-month aneurysm progression (mean difference at 18 months: -0.24 mm (95% CI: -0.90-0.45, P = NS). CONCLUSIONS ACE inhibition quenches multiple aspects of vascular inflammation in AAA. However, this does not translate into reduced aneurysm growth. TRIAL REGISTRATION Nederlands Trial Register 1345.
Collapse
Affiliation(s)
- Kim E. Kortekaas
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
- Division of Vascular Surgery, Stanford Medical School, Stanford, California, United States of America
| | - C. Arnoud Meijer
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan Willem Hinnen
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
- Department of Vascular Surgery, Jeroen Bosch Ziekenhuis, ‘s Hertogenbosch, The Netherlands
| | - Ronald L. Dalman
- Division of Vascular Surgery, Stanford Medical School, Stanford, California, United States of America
| | - Baohui Xu
- Division of Vascular Surgery, Stanford Medical School, Stanford, California, United States of America
| | - Jaap F. Hamming
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan H. Lindeman
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
81
|
A lovastatin-elicited genetic program inhibits M2 macrophage polarization and enhances T cell infiltration into spontaneous mouse mammary tumors. Oncotarget 2014; 4:2288-301. [PMID: 24317954 PMCID: PMC3926827 DOI: 10.18632/oncotarget.1376] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Beyond their ability to inhibit cholesterol biosynthesis, the statins have pleiotropic effects that include anti-inflammatory and immunomodulatory activities. Statins could have clinical utility, alone or in combination with other chemotherapeutics, in the treatment of cancer. The mechanisms that underlie the anti-tumor activity of the statins are nonetheless poorly defined. No studies have analyzed how they alter the tumor-associated leukocyte infiltrate, a central factor that influences tumor stroma and cancer evolution. Here we used HER2/neu transgenic (Tg-neu) mice to analyze the effect of lovastatin (Lov) on the inflammatory reaction of spontaneous mammary tumors. Lov treatment of tumor-bearing Tg-neu mice did not alter growth of established tumors, but significantly reduced the number of new oncogenic lesions in these mice. Moreover, Lov inhibited the growth of newly implanted Tg-neu tumors in immunocompetent but not in immunodeficient mice. We found that Lov enhanced tumor infiltration by effector T cells, and reduced the number of immunosuppressive and pro-angiogenic M2-like tumor-associated macrophages (TAM). Concomitantly, the drug improved the structure and function of the tumor vasculature, measured as enhanced tumor oxygenation and penetration of cytotoxic drugs. Microarray analysis identified a Lov-elicited genetic program in Tg-neu tumors that might explain these effects; we observed Lov-induced downregulation of placental growth factor, which triggers aberrant angiogenesis and M2-like TAM polarization. Our results identify a role for lovastatin in the shaping and re-education of the inflammatory infiltrate in tumors, with functional consequences in angiogenesis and antitumor immunity.
Collapse
|
82
|
Drolz A, Horvatits T, Michl B, Roedl K, Schellongowski P, Holzinger U, Zauner C, Heinz G, Madl C, Trauner M, Fuhrmann V. Statin therapy is associated with reduced incidence of hypoxic hepatitis in critically ill patients. J Hepatol 2014; 60:1187-93. [PMID: 24509409 DOI: 10.1016/j.jhep.2014.01.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Revised: 01/23/2014] [Accepted: 01/24/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Hypoxic hepatitis (HH) is a frequent and life-threatening complication associated with states of oxygen depletion in critically ill patients. Ischemia and reperfusion contribute to liver injury in HH. Experimental data suggest beneficial effects of statins in hepatic ischemia/reperfusion injury. This study was conducted to investigate whether statin treatment prior to intensive care unit (ICU) admission affects incidence rates and severity of HH. METHODS Eight hundred fifty-one patients admitted consecutively to three medical ICUs between December 2008 and December 2009 were prospectively screened for new occurrence of HH within 48 h following ICU admission. Statin treatment prior to ICU admission was assessed. 28-day-, 90-day-, and 1-year-survival as well as new-onset of complications in HH patients were prospectively documented. RESULTS Eighty-seven patients (10%) developed HH. Statin treatment prior to ICU admission was significantly associated with decreased incidence of HH within 48 h after ICU admission in the multivariate analysis (adjusted OR=0.42 (95% CI 0.19-0.95); p<0.05). Cardiogenic shock (p<0.001), septic shock (p<0.001) and active alcohol consumption (p<0.01) were identified as independent risk factors for development of HH. 28-day-, 90-day-, and 1-year-mortality rates in HH were 58%, 67%, and 74%, respectively. Statins were associated with improved 28-day-survival in the total study cohort (p<0.05), but did not affect 90-day- and 1-year-mortality, respectively. CONCLUSIONS Cardiogenic shock, septic shock, and active alcohol consumption were independent factors predisposing patients to new onset of HH. Statin treatment prior to ICU admission was the only protective factor regarding the new occurrence of HH in critically ill patients.
Collapse
Affiliation(s)
- Andreas Drolz
- Intensive Care Unit 13 H1, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria
| | - Thomas Horvatits
- Intensive Care Unit 13 H1, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria
| | - Barbara Michl
- Intensive Care Unit 13 H1, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria
| | - Kevin Roedl
- Intensive Care Unit 13 H1, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria
| | - Peter Schellongowski
- Intensive Care Unit 13 I2, Department of Oncology and Infectious Diseases, Division of Internal Medicine I, Medical University of Vienna, Austria
| | - Ulrike Holzinger
- Intensive Care Unit 13 H1, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria
| | - Christian Zauner
- Intensive Care Unit 13 H1, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria
| | - Gottfried Heinz
- Intensive Care Unit 13 H3, Department of Cardiology, Division of Internal Medicine II, Medical University of Vienna, Austria
| | - Christian Madl
- Department of Gastroenterology and Hepatology, KA Rudolfstiftung, Austria
| | - Michael Trauner
- Intensive Care Unit 13 H1, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria
| | - Valentin Fuhrmann
- Intensive Care Unit 13 H1, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Austria.
| |
Collapse
|
83
|
The ω-3 polyunsaturated fatty acid, eicosapentaenoic acid, attenuates abdominal aortic aneurysm development via suppression of tissue remodeling. PLoS One 2014; 9:e96286. [PMID: 24798452 PMCID: PMC4010435 DOI: 10.1371/journal.pone.0096286] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 04/06/2014] [Indexed: 01/24/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a prevalent vascular disease that can progressively enlarge and rupture with a high rate of mortality. Inflammation and active remodeling of the aortic wall have been suggested to be critical in its pathogenesis. Meanwhile, ω-3 polyunsaturated fatty acids such as eicosapentaenoic acid (EPA) are known to reduce cardiovascular events, but its role in AAA management remains unclear. Here, we show that EPA can attenuate murine CaCl2-induced AAA formation. Aortas from BALB/c mice fed an EPA-diet appeared less inflamed, were significantly smaller in diameter compared to those from control-diet-fed mice, and had relative preservation of aortic elastic lamina. Interestingly, CT imaging also revealed markedly reduced calcification of the aortas after EPA treatment. Mechanistically, MMP2, MMP9, and TNFSF11 levels in the aortas were reduced after EPA treatment. Consistent with this finding, RAW264.7 macrophages treated with EPA showed attenuated Mmp9 levels after TNF-α simulation. These results demonstrate a novel role of EPA in attenuating AAA formation via the suppression of critical remodeling pathways in the pathogenesis of AAAs, and raise the possibility of using EPA for AAA prevention in the clinical setting.
Collapse
|
84
|
Pellegrin M, Bouzourène K, Poitry-Yamate C, Mlynarik V, Feihl F, Aubert JF, Gruetter R, Mazzolai L. Experimental peripheral arterial disease: new insights into muscle glucose uptake, macrophage, and T-cell polarization during early and late stages. Physiol Rep 2014; 2:e00234. [PMID: 24744903 PMCID: PMC3966252 DOI: 10.1002/phy2.234] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 01/20/2014] [Indexed: 01/28/2023] Open
Abstract
Peripheral arterial disease (PAD) is a common disease with increasing prevalence, presenting with impaired walking ability affecting patient's quality of life. PAD epidemiology is known, however, mechanisms underlying functional muscle impairment remain unclear. Using a mouse PAD model, aim of this study was to assess muscle adaptive responses during early (1 week) and late (5 weeks) disease stages. Unilateral hindlimb ischemia was induced in ApoE−/− mice by iliac artery ligation. Ischemic limb perfusion and oxygenation (Laser Doppler imaging, transcutaneous oxygen pressure assessments) significantly decreased during early and late stage compared to pre‐ischemia, however, values were significantly higher during late versus early phase. Number of arterioles and arteriogenesis‐linked gene expression increased at later stage. Walking ability, evaluated by forced and voluntary walking tests, remained significantly decreased both at early and late phase without any significant improvement. Muscle glucose uptake ([18F]fluorodeoxyglucose positron emission tomography) significantly increased during early ischemia decreasing at later stage. Gene expression analysis showed significant shift in muscle M1/M2 macrophages and Th1/Th2 T cells balance toward pro‐inflammatory phenotype during early ischemia; later, inflammatory state returned to neutrality. Muscular M1/M2 shift inhibition by a statin prevented impaired walking ability in early ischemia. High‐energy phosphate metabolism remained unchanged (31‐Phosphorus magnetic resonance spectroscopy). Results show that rapid transient muscular inflammation contributes to impaired walking capacity while increased glucose uptake may be a compensatory mechanisms preserving immediate limb viability during early ischemia in a mouse PAD model. With time, increased ischemic limb perfusion and oxygenation assure muscle viability although not sufficiently to improve walking impairment. Subsequent decreased muscle glucose uptake may partly contribute to chronic walking impairment. Early inflammation inhibition and/or late muscle glucose impairment prevention are promising strategies for PAD management. e00234 Mechanisms responsible for functional muscle impairment in peripheral artery disease patients remain unknown. Our results show that both pro‐inflammatory T cells and macrophages are implicated in early peripheral ischemia whereas late ischemia is associated with impaired muscle glucose uptake in a mouse model of peripheral arterial disease (PAD).
Collapse
Affiliation(s)
- Maxime Pellegrin
- Division of Angiology, University Hospital of Lausanne, Lausanne, Switzerland
| | - Karima Bouzourène
- Division of Angiology, University Hospital of Lausanne, Lausanne, Switzerland
| | - Carole Poitry-Yamate
- Centre d'Imagerie Biomédicale, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Vladimir Mlynarik
- Centre d'Imagerie Biomédicale, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - François Feihl
- Division of Clinical Pathophysiology, University Hospital of Lausanne, Lausanne, Switzerland
| | | | - Rolf Gruetter
- Centre d'Imagerie Biomédicale, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Lucia Mazzolai
- Division of Angiology, University Hospital of Lausanne, Lausanne, Switzerland
| |
Collapse
|
85
|
Patel JM, Thickett DR, Gao F, Sapey E. Statins for sepsis: distinguishing signal from the noise when designing clinical trials. Am J Respir Crit Care Med 2013; 188:874. [PMID: 24083863 DOI: 10.1164/rccm.201302-0392le] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
86
|
Wang L, Fu Y, Peng J, Wu D, Yu M, Xu C, Wang Q, Tao L. Simvastatin-induced up-regulation of gap junctions composed of connexin 43 sensitize Leydig tumor cells to etoposide: An involvement of PKC pathway. Toxicology 2013; 312:149-57. [DOI: 10.1016/j.tox.2013.08.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 08/05/2013] [Accepted: 08/13/2013] [Indexed: 02/02/2023]
|
87
|
Kuivaniemi H, Sakalihasan N, Lederle FA, Jones GT, Defraigne JO, Labropoulos N, Legrand V, Michel JB, Nienaber C, Radermecker MA, Elefteriades JA. New Insights Into Aortic Diseases: A Report From the Third International Meeting on Aortic Diseases (IMAD3). AORTA (STAMFORD, CONN.) 2013; 1:23-39. [PMID: 26798669 PMCID: PMC4682695 DOI: 10.12945/j.aorta.2013.13.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 03/08/2013] [Indexed: 12/11/2022]
Abstract
The current state of research and treatment on aortic diseases was discussed in the "3rd International Meeting on Aortic Diseases" (IMAD3) held on October 4-6, 2012, in Liège, Belgium. The 3-day meeting covered a wide range of topics related to thoracic aortic aneurysms and dissections, abdominal aortic aneurysms, and valvular diseases. It brought together clinicians and basic scientists and provided an excellent opportunity to discuss future collaborative research projects for genetic, genomics, and biomarker studies, as well as clinical trials. Although great progress has been made in the past few years, there are still a large number of unsolved questions about aortic diseases. Obtaining answers to the key questions will require innovative, interdisciplinary approaches that integrate information from epidemiological, genetic, molecular biology, and bioengineering studies on humans and animal models. It is more evident than ever that multicenter collaborations are needed to accomplish these goals.
Collapse
Affiliation(s)
- Helena Kuivaniemi
- Sigfried and Janet Weis Center for Research, Geisinger Clinic, Danville, Pennsylvania
| | | | - Frank A. Lederle
- Minneapolis Center for Epidemiological and Clinical Research, Department of Medicine (III-0), VA Medical Center, Minneapolis, Minnesota
| | | | | | - Nicos Labropoulos
- Department of Surgery, Stony Brook University Medical Center, Stony Brook, New York
| | - Victor Legrand
- Cardiology Departments, University Hospital of Liège, CHU, Liège, Belgium
| | | | | | | | | |
Collapse
|