151
|
Spiteller G, Afzal M. The action of peroxyl radicals, powerful deleterious reagents, explains why neither cholesterol nor saturated fatty acids cause atherogenesis and age-related diseases. Chemistry 2014; 20:14928-45. [PMID: 25318456 DOI: 10.1002/chem.201404383] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cells respond to alterations in their membrane structure by activating hydrolytic enzymes. Thus, polyunsaturated fatty acids (PUFAs) are liberated. Free PUFAs react with molecular oxygen to give lipid hydroperoxide molecules (LOOHs). In case of severe cell injury, this physiological reaction switches to the generation of lipid peroxide radicals (LOO(·)). These radicals can attack nearly all biomolecules such as lipids, carbohydrates, proteins, nucleic acids and enzymes, impairing their biological functions. Identical cell responses are triggered by manipulation of food, for example, heating/grilling and particularly homogenization, representing cell injury. Cholesterol as well as diets rich in saturated fat have been postulated to accelerate the risk of atherosclerosis while food rich in unsaturated fatty acids has been claimed to lower this risk. However, the fact is that LOO(·) radicals generated from PUFAs can oxidize cholesterol to toxic cholesterol oxides, simulating a reduction in cholesterol level. In this review it is shown how active LOO(·) radicals interact with biomolecules at a speed transcending usual molecule-molecule reactions by several orders of magnitude. Here, it is explained how functional groups are fundamentally transformed by an attack of LOO(·) with an obliteration of essential biomolecules leading to pathological conditions. A serious reconsideration of the health and diet guidelines is required.
Collapse
Affiliation(s)
- Gerhard Spiteller
- University of Bayreuth, Universitätsstr. 30, 95445 Bayreuth (Germany).
| | | |
Collapse
|
152
|
Coronary atheroma composition and its association with segmental endothelial dysfunction in non-ST segment elevation myocardial infarction: novel insights with radiofrequency (iMAP) intravascular ultrasonography. Int J Cardiovasc Imaging 2014; 31:247-57. [DOI: 10.1007/s10554-014-0545-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 09/29/2014] [Indexed: 01/23/2023]
|
153
|
Huang X, Wang F, Chen W, Wang N, Chen Y, Sun L. DaoTan decoction (DTD) inhibits tumor necrosis factor-α (TNF-α)-induced expression of intercellular adhesion molecule-1 (ICAM-1), p53 and p21, in human umbilical vein endothelia cells (HUVECs). PHARMACEUTICAL BIOLOGY 2014; 52:1320-1326. [PMID: 25026345 DOI: 10.3109/13880209.2014.891141] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
CONTEXT DTD is a Chinese herb prescription used for centuries to treat atherosclerosis or dizziness. Previous studies show that DTD could inhibit ICAM-1 expression induced by TNF-α. However, its mechanism has never been clearly described. OBJECTIVE To examine the hypothesis that DTD might inhibit TNF-α-induced ICAM-1 expression through regulating the expression of p53 and p21. MATERIALS AND METHODS The rats were orally treated with DTD for 3 d (2.3 g/kg per day), and then the serum was collected. HUVECs were cultured and stimulated by TNF-α with or without DTD serum (5, 10, and 20%). The expression of ICAM-1 mRNA was examined by RT-PCR and the expression of p53 and p21 was examined by western blot analysis. RESULTS The ICAM-1 mRNA levels induced by TNF-α were significantly reduced from 23 to 47%, and the expression of p53 and p21 mRNA levels were significantly reduced from 13 to 43% and 14 to 42%, as the concentration of DTD serum increased. In western blot, TNF-α-induced the expression of p53 and was inhibited from 15 to 53%, by DTD serum in a concentration-dependent manner. TNF-α-induced expression of p21 was inhibited from 2 to 37%, by DTD serum in a concentration-dependent manner. DISCUSSION AND CONCLUSION DTD has a function of "dissolving phlegm", thus it is chosen for the treatment of atherosclerosis. This study demonstrated that DTD could significantly inhibit the expression of ICAM-1, p53 and p21, which are important factors of atherosclerosis. Therefore, the present study indicates the pharmacological basis for treatment of atherosclerosis with DTD.
Collapse
Affiliation(s)
- Xiaobo Huang
- Department of Chinese Medicine, Xuanwu Hospital, Capital Medical University , Beijing , China
| | | | | | | | | | | |
Collapse
|
154
|
Sakamoto S, Tsuruda T, Hatakeyama K, Imamura T, Asada Y, Kitamura K. Impact of age-dependent adventitia inflammation on structural alteration of abdominal aorta in hyperlipidemic mice. PLoS One 2014; 9:e105739. [PMID: 25153991 PMCID: PMC4143271 DOI: 10.1371/journal.pone.0105739] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 07/28/2014] [Indexed: 11/23/2022] Open
Abstract
Background The adventitia is suggested to contribute to vascular remodeling; however, the site-selective inflammatory responses in association with the development of atherosclerosis remain to be elucidated. Methods and Results Wild-type or apolipoprotein E knockout male C57BL/6J background mice were fed standard chow for 16, 32, and 52 weeks, and the morphology of the aortic arch, descending aorta, and abdominal aorta was compared. Atheromatous plaque formation progressed with age, particularly in the aortic arch and abdominal aorta but not in the descending aorta. In addition, we found that the numbers of macrophages, T-lymphocytes, and microvessels, assessed by anti-F4/80, CD3, and CD31 antibodies, were higher in the adventitia of the abdominal aorta at 52 weeks. These numbers were positively correlated with plaque formation, but negatively correlated with elastin content, resulting in the enlargement of the total vessel area. In aortic tissues, interleukin-6 levels increased in the atheromatous plaque with age, whereas the level of regulated on activation, normal T cell expressed and secreted (RANTES) increased with age, and compared with other sites, it was particularly distributed in inflammatory cells in the adventitia of the abdominal aorta. Conclusion This study suggests that adventitial inflammation contributes to the age-dependent structural alterations, and that the activation/inactivation of cytokines/chemokines is involved in the process.
Collapse
Affiliation(s)
- Sumiharu Sakamoto
- Department of Internal Medicine, Circulatory and Body Fluid Regulation, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Toshihiro Tsuruda
- Department of Internal Medicine, Circulatory and Body Fluid Regulation, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Kinta Hatakeyama
- Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Takuroh Imamura
- Department of Internal Medicine, Koga General Hospital, Miyazaki, Japan
| | - Yujiro Asada
- Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Kazuo Kitamura
- Department of Internal Medicine, Circulatory and Body Fluid Regulation, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| |
Collapse
|
155
|
Wiberg B, Lind PM, Lind L. Serum levels of monobenzylphthalate (MBzP) is related to carotid atherosclerosis in the elderly. ENVIRONMENTAL RESEARCH 2014; 133:348-352. [PMID: 25036990 DOI: 10.1016/j.envres.2014.06.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 06/04/2014] [Accepted: 06/11/2014] [Indexed: 06/03/2023]
Abstract
BACKGROUND AND OBJECTIVE Background exposure to environmental contaminants has recently emerged as a new risk factor for cardiovascular disease in general and to atheroclerosis in particular. This cross-sectional study was performed to evaluate if serum concentrations of the phthalate metabolite monobenzylphthalate (MBzP) are related to atheroclerosis in the carotid arteries. METHODS In the population-based Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS) study (1003 subjects all aged 70) the prevalence of overt plaques and echogenicity (gray-scale median, GSM) of carotid artery plaques were recorded by ultrasound in both of the carotid arteries. The intima-media thickness (IMT) and echogenicity (IM-GSM) of the intima-media complex were also measured. The phthalate metabolite MBzP was analyzed in serum by a liquid chromatograph/tandem mass spectrometer. RESULTS The circulating level of the phthalate MBzP was related to intima-media thickness (IMT) when adjusting for gender, blood pressure, body mass index (BMI), waist circumference, HDL- and LDL-cholesterol, serum triglycerides, blood glucose and smoking (p=0.034). High levels of MBzP were also strongly associated with an echogenic IM-GSM and plaque GSM (p=0.0001 for both outcomes after adjustment) but not to plaque prevalence (p=0.42). CONCLUSION The phthalate metabolite MBzP was strongly related to the echogenicity of intima-media and plaques and also to IMT, independently of traditional CV risk factors. This suggests a role for the phthalate MBzP in the development of atherosclerosis.
Collapse
Affiliation(s)
- Bernice Wiberg
- Department of Public Health and Caring Sciences/Geriatrics, Uppsala University Hospital, SE-751 85 Uppsala, Sweden.
| | - P Monica Lind
- Occupational and Environmental Medicine, Uppsala University, Uppsala, Sweden
| | - Lars Lind
- Department of Medicine, Uppsala University, Uppsala, Sweden
| |
Collapse
|
156
|
|
157
|
Prüfer J, Schuchardt M, Tölle M, Prüfer N, Höhne M, Zidek W, van der Giet M. Harmful effects of the azathioprine metabolite 6-mercaptopurine in vascular cells: induction of mineralization. PLoS One 2014; 9:e101709. [PMID: 25029363 PMCID: PMC4100760 DOI: 10.1371/journal.pone.0101709] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 06/10/2014] [Indexed: 12/31/2022] Open
Abstract
Vascular mineralization contributes to the high cardiovascular morbidity and mortality in patients who suffer from chronic kidney disease and in individuals who have undergone solid organ transplantation. The immunosuppressive regimen used to treat these patients appears to have an impact on vascular alterations. The effect of 6-mercaptopurine (6-MP) on vascular calcification has not yet been determined. This study investigates the effect of 6-MP on vascular mineralization by the induction of trans-differentiation of rat vascular smooth muscle cells in vitro. 6-MP not only induces the expression of osteo-chondrocyte-like transcription factors and proteins but also activates alkaline phosphatase enzyme activity and produces calcium deposition in in vitro and ex vivo models. These processes are dependent on 6-MP-induced production of reactive oxygen species, intracellular activation of mitogen-activated kinases and phosphorylation of the transcription factor Cbfa1. Furthermore, the metabolic products of 6-MP, 6-thioguanine nucleotides and 6-methyl-thio-inosine monophosphate have major impacts on cellular calcification. These data provide evidence for a possible harmful effect of the immunosuppressive drug 6-MP in vascular diseases, such as arteriosclerosis.
Collapse
Affiliation(s)
- Jasmin Prüfer
- Charité – Universitaetsmedizin Berlin; Charité Centrum 13, Department of Nephrology Campus Benjamin Franklin, Berlin, Germany
- Freie Universitaet Berlin, Fachbereich Biochemie, Chemie, Berlin, Germany
| | - Mirjam Schuchardt
- Charité – Universitaetsmedizin Berlin; Charité Centrum 13, Department of Nephrology Campus Benjamin Franklin, Berlin, Germany
| | - Markus Tölle
- Charité – Universitaetsmedizin Berlin; Charité Centrum 13, Department of Nephrology Campus Benjamin Franklin, Berlin, Germany
| | - Nicole Prüfer
- Charité – Universitaetsmedizin Berlin; Charité Centrum 13, Department of Nephrology Campus Benjamin Franklin, Berlin, Germany
- Universitaet Potsdam, Department of Nutrition Science, Potsdam, Germany
| | | | - Walter Zidek
- Charité – Universitaetsmedizin Berlin; Charité Centrum 13, Department of Nephrology Campus Benjamin Franklin, Berlin, Germany
| | - Markus van der Giet
- Charité – Universitaetsmedizin Berlin; Charité Centrum 13, Department of Nephrology Campus Benjamin Franklin, Berlin, Germany
- * E-mail:
| |
Collapse
|
158
|
Puri R, Nicholls SJ, Brennan DM, Andrews J, King KL, Liew GY, Carbone A, Copus B, Nelson AJ, Kapadia SR, Tuzcu EM, Beltrame JF, Worthley SG, Worthley MI. Left main coronary arterial endothelial function and heterogenous segmental epicardial vasomotor reactivity in vivo: novel insights with intravascular ultrasonography. Eur Heart J Cardiovasc Imaging 2014; 15:1270-80. [PMID: 25024410 DOI: 10.1093/ehjci/jeu133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS While the relationship between epicardial coronary vasomotor reactivity and cardiovascular events is well established, this observation has yet to be evaluated within the left main coronary artery (LMCA) in humans in vivo. Our aims were to test the endothelium-dependent vasomotor properties of the LMCA, and to compare these responses to downstream epicardial segments. METHODS AND RESULTS Thirty patients referred for coronary angiography underwent intracoronary (IC) salbutamol provocation during intravascular ultrasound imaging within a non-critically diseased, left-sided conduit vessel. Macrovascular vasomotor response [change in average lumen area (LA) at baseline and following 5 min of 0.30 µg/min IC salbutamol] and percent atheroma volume (PAV) were evaluated in 30 LMCA, 42 proximal, 109 mid, and 132 distal epicardial coronary segments. In comparison with all other segments, the LMCA had the greatest lumen and vessel areas (P < 0.001), yet the proximal epicardial segments contained the greatest PAV (P < 0.02). The mid and distal epicardial segments displayed significant endothelium-dependent vasodilatation from baseline (P = 0.017 and <0.001, respectively); however, the proximal epicardial and LMCA segments did not (P = 0.45 and 0.16, respectively). Significant segmental vasomotor heterogeneity was noted in all 30 patients, with opposing vasomotor responses between adjacent LMCA and epicardial segments. Across all segments, baseline LA inversely correlated with the % change in LA (r = -0.16, P = 0.0005). CONCLUSION Endothelium-dependent vasomotor reactivity is heterogenous within the conduit coronary system. Vascular dynamic responses were less prominent in the larger calibre LMCA and proximal epicardial segments. This may, in part, relate to higher shear stress in smaller, distal segments and yet also may explain the propensity for culprit plaques to cluster proximally.
Collapse
Affiliation(s)
- Rishi Puri
- Discipline of Medicine, University of Adelaide, Adelaide, Australia Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH, USA C5Research, Cleveland Clinic, Cleveland, OH, USA
| | - Stephen J Nicholls
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia Cardiovascular Investigation Unit, Royal Adelaide Hospital, Adelaide, Australia
| | | | - Jordan Andrews
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| | | | - Gary Y Liew
- Discipline of Medicine, University of Adelaide, Adelaide, Australia
| | - Angelo Carbone
- Discipline of Medicine, University of Adelaide, Adelaide, Australia
| | - Barbara Copus
- Cardiovascular Investigation Unit, Royal Adelaide Hospital, Adelaide, Australia
| | - Adam J Nelson
- Discipline of Medicine, University of Adelaide, Adelaide, Australia
| | - Samir R Kapadia
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Emin Murat Tuzcu
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - John F Beltrame
- Discipline of Medicine, University of Adelaide, Adelaide, Australia
| | - Stephen G Worthley
- Discipline of Medicine, University of Adelaide, Adelaide, Australia Cardiovascular Investigation Unit, Royal Adelaide Hospital, Adelaide, Australia Department of Medicine, Cardiovascular Research Centre, Royal Adelaide Hospital, Level 6, Theatre Block, North Terrace, Adelaide, SA 5000, Australia
| | - Matthew I Worthley
- Discipline of Medicine, University of Adelaide, Adelaide, Australia Cardiovascular Investigation Unit, Royal Adelaide Hospital, Adelaide, Australia Department of Medicine, Cardiovascular Research Centre, Royal Adelaide Hospital, Level 6, Theatre Block, North Terrace, Adelaide, SA 5000, Australia
| |
Collapse
|
159
|
Vorpahl M, Schönhofer-Merl S, Michaelis C, Flotho A, Melchior F, Wessely R. The Ran GTPase-activating protein (RanGAP1) is critically involved in smooth muscle cell differentiation, proliferation and migration following vascular injury: implications for neointima formation and restenosis. PLoS One 2014; 9:e101519. [PMID: 24988324 PMCID: PMC4079658 DOI: 10.1371/journal.pone.0101519] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 06/07/2014] [Indexed: 12/12/2022] Open
Abstract
Differentiation and dedifferentiation, accompanied by proliferation play a pivotal role for the phenotypic development of vascular proliferative diseases (VPD), such as restenosis. Increasing evidence points to an essential role of regulated nucleoporin expression in the choice between differentiation and proliferation. However, whether components of the Ran GTPase cycle, which is of pivotal importance for both nucleocytoplasmic transport and for mitotic progression, are subject to similar regulation in VPD is currently unknown. Here, we show that differentiation of human coronary artery smooth muscle cell (CASMC) to a contractile phenotype by stepwise serum depletion leads to significant reduction of RanGAP1 protein levels. The inverse event, dedifferentiation of cells, was assessed in the rat carotid artery balloon injury model, a well-accepted model for neointima formation and restenosis. As revealed by temporospatial analysis of RanGAP1 expression, neointima formation in rat carotid arteries was associated with a significant upregulation of RanGAP1 expression at 3 and 7 days after balloon injury. Of note, neointimal cells located at the luminal surface revealed persistent RanGAP1 expression, as opposed to cells in deeper layers of the neointima where RanGAP1 expression was less or not detectable at all. To gain first evidence for a direct influence of RanGAP1 levels on differentiation, we reduced RanGAP1 in human coronary artery smooth muscle cells by siRNA. Indeed, downregulation of the essential RanGAP1 protein by 50% induced a differentiated, spindle-like smooth muscle cell phenotype, accompanied by an upregulation of the differentiation marker desmin. Reduction of RanGAP1 levels also resulted in a reduction of mitogen induced cellular migration and proliferation as well as a significant upregulation of the cyclin-dependent kinase inhibitor p27KIP1, without evidence for cellular necrosis. These findings suggest that RanGAP1 plays a critical role in smooth muscle cell differentiation, migration and proliferation in vitro and in vivo. Appropriate modulation of RanGAP1 expression may thus be a strategy to modulate VPD development such as restenosis.
Collapse
Affiliation(s)
- Marc Vorpahl
- Department of Cardiology, HELIOS Klinikum Wuppertal, University Witten/Herdecke, Witten, Germany
- * E-mail: (MV); (RW)
| | - Sabine Schönhofer-Merl
- Deutsches Herzzentrum Muenchen and 1. Medizinische Klinik, Klinikum rechts der Isar, Technische Universitaet Muenchen, Muenchen, Germany
| | - Cornelia Michaelis
- Deutsches Herzzentrum Muenchen and 1. Medizinische Klinik, Klinikum rechts der Isar, Technische Universitaet Muenchen, Muenchen, Germany
| | - Annette Flotho
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ - ZMBH Alliance, Heidelberg, Germany
| | - Frauke Melchior
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ - ZMBH Alliance, Heidelberg, Germany
| | - Rainer Wessely
- Deutsches Herzzentrum Muenchen and 1. Medizinische Klinik, Klinikum rechts der Isar, Technische Universitaet Muenchen, Muenchen, Germany
- Zentrum fuer Herz-Gefaess-Lungenmedizin, Cologne, Germany
- * E-mail: (MV); (RW)
| |
Collapse
|
160
|
Inhibition of vascular calcification by block of intermediate conductance calcium-activated potassium channels with TRAM-34. Pharmacol Res 2014; 85:6-14. [DOI: 10.1016/j.phrs.2014.04.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 04/25/2014] [Accepted: 04/27/2014] [Indexed: 01/08/2023]
|
161
|
Role of small GTPase protein Rac1 in cardiovascular diseases: development of new selective pharmacological inhibitors. J Cardiovasc Pharmacol 2014; 62:425-35. [PMID: 23921306 DOI: 10.1097/fjc.0b013e3182a18bcc] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A pathway-based genome-wide association analysis has recently identified Rac1 as one of the biologically important gene in coronary heart diseases. The role of the small GTPase Rac1 in cardiac hypertrophy and atherosclerosis has also been documented in clinical studies with the HMG-CoA reductase inhibitors and in in vitro and in vivo settings using transgenic and knockout mice. Thus, Rac1 has emerged as a new pharmacological target for the treatment of cardiovascular diseases. The activation state of Rac1 depends on the release of guanosine diphosphate and the binding of guanosine triphosphate. This cycling is regulated by the guanine nucleotide exchange factors, as activators, and by the GTPase-activating proteins. Three categories of selective Rac1 inhibitors have been developed affecting different steps of this pathway: antagonists of Rac1-guanine nucleotide exchange factor interaction, allosteric inhibitors of nucleotide binding to Rac1, and antagonists of Rac1-mediated NADPH oxidase activity. These chemical compounds have shown to selectively inhibit Rac1 activation in cultured cell lines without affecting the homologous proteins RhoA and Cdc42. Moreover, pioneer studies have been conducted with Rac1 inhibitors in in vivo experimental models of cardiovascular diseases with encouraging results. The present review summarizes the current knowledge of the role of Rac1 in cardiovascular diseases and the pharmacological approaches that have been developed to selectively inhibit its function.
Collapse
|
162
|
|
163
|
Mohanta SK, Yin C, Peng L, Srikakulapu P, Bontha V, Hu D, Weih F, Weber C, Gerdes N, Habenicht AJ. Artery Tertiary Lymphoid Organs Contribute to Innate and Adaptive Immune Responses in Advanced Mouse Atherosclerosis. Circ Res 2014; 114:1772-87. [PMID: 24855201 DOI: 10.1161/circresaha.114.301137] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tertiary lymphoid organs emerge in tissues in response to nonresolving inflammation. Recent research characterized artery tertiary lymphoid organs in the aorta adventitia of aged apolipoprotein E–deficient mice. The atherosclerosis-associated lymphocyte aggregates are organized into distinct compartments, including separate T-cell areas harboring conventional, monocyte-derived, lymphoid, and plasmacytoid dendritic cells, as well as activated T-cell effectors and memory cells; B-cell follicles containing follicular dendritic cells in activated germinal centers; and peripheral niches of plasma cells. Artery tertiary lymphoid organs show marked neoangiogenesis, aberrant lymphangiogenesis, and extensive induction of high endothelial venules. Moreover, newly formed lymph node–like conduits connect the external lamina with high endothelial venules in T-cell areas and also extend into germinal centers. Mouse artery tertiary lymphoid organs recruit large numbers of naïve T cells and harbor lymphocyte subsets with opposing activities, including CD4
+
and CD8
+
effector and memory T cells, natural and induced CD4
+
regulatory T cells, and memory B cells at different stages of differentiation. These data suggest that artery tertiary lymphoid organs participate in primary immune responses and organize T- and B-cell autoimmune responses in advanced atherosclerosis. In this review, we discuss the novel concept that pro- and antiatherogenic immune responses toward unknown arterial wall–derived autoantigens may be organized by artery tertiary lymphoid organs and that disruption of the balance between pro- and antiatherogenic immune cell subsets may trigger clinically overt atherosclerosis.
Collapse
Affiliation(s)
- Sarajo Kumar Mohanta
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Changjun Yin
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Li Peng
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Prasad Srikakulapu
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Vineela Bontha
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Desheng Hu
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Falk Weih
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Christian Weber
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Norbert Gerdes
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| | - Andreas J.R. Habenicht
- From the Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (S.K.M., C.Y., C.W., N.G., A.J.R.H.); Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena, Germany (L.P., P.S., V.B., F.W.); and Institute of Molecular Immunology, Helmholtz Center Munich, Neuherberg, Germany (D.H.)
| |
Collapse
|
164
|
Mori T, Koyama Y, Maeda N, Nakamura Y, Fujishima Y, Matsuda K, Funahashi T, Shimada S, Shimomura I. Ultrastructural localization of adiponectin protein in vasculature of normal and atherosclerotic mice. Sci Rep 2014; 4:4895. [PMID: 24809933 PMCID: PMC4013939 DOI: 10.1038/srep04895] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 04/17/2014] [Indexed: 11/25/2022] Open
Abstract
Adiponectin, adipose-specific secretory protein, abundantly circulates in bloodstream and its concentration is around 1000-fold higher than that of other cytokines and hormones. Hypoadiponectinemia is a risk factor for atherosclerosis. There is little or no information on ultrastructural localization of adiponectin in the vasculature. Herein we investigated the localization of vascular adiponectin in the aorta using the immunoelectron microscopic technique. In wild-type (WT) mice, adiponectin was mainly detected on the luminal surface membrane of endothelial cells (ECs) and also found intracellularly in the endocytic vesicles of ECs. In the atherosclerotic lesions of apolipoprotein E-knockout (ApoE-KO) mice, adiponectin was detected in ECs, on the cell surface membrane of synthetic smooth muscle cells, and on the surface of monocytes adherent to ECs. Changes in adiponectin localization within the wall of the aorta may provide novel insight into the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Takuya Mori
- 1] Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan 565-0871 [2]
| | - Yoshihisa Koyama
- 1] Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan 565-0871 [2]
| | - Norikazu Maeda
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan 565-0871
| | - Yukiko Nakamura
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan 565-0871
| | - Yuya Fujishima
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan 565-0871
| | - Keisuke Matsuda
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan 565-0871
| | - Tohru Funahashi
- 1] Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan 565-0871 [2] Department of Metabolism and Atherosclerosis, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan 565-0871
| | - Shoichi Shimada
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan 565-0871
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan 565-0871
| |
Collapse
|
165
|
Kuloglu T, Aydin S, Eren MN, Yilmaz M, Sahin I, Kalayci M, Sarman E, Kaya N, Yilmaz OF, Turk A, Aydin Y, Yalcin MH, Uras N, Gurel A, Ilhan S, Gul E, Aydin S. Irisin: a potentially candidate marker for myocardial infarction. Peptides 2014; 55:85-91. [PMID: 24576483 DOI: 10.1016/j.peptides.2014.02.008] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 02/06/2014] [Accepted: 02/07/2014] [Indexed: 12/30/2022]
Abstract
Myocardial infarction (MI) causes energy depletion through imbalance between coronary blood supply and myocardial demand. Irisin produced by the heart reduces ATP production by increasing heat generation. Energy depletion affects irisin concentration in circulation and cardiac tissues, suggesting an association with MI. We examined: (1) irisin expression immunohistochemically in rat heart, skeletal muscle, kidney and liver in isoproterenol (ISO)-induced MI, and (2) serum irisin concentration by ELISA. Rats were randomly allocated into 6 groups (n=6), (i) control, (ii) ISO (1h), (iii) ISO (2h), (iv) ISO (4h), (v) ISO (6h), and (vi) ISO (24h), 200mg ISO in each case. Rats were decapitated and the blood and tissues collected for irisin analysis. Blood was centrifuged at 1792 g for 5 min. Tissues were washed with saline and fixed in 10% formalin for histology. Serum irisin levels gradually decreased from 1h to 24h in MI rats compared with controls, the minimum being at 2h, increasing again after 6h. Cardiac muscle cells, glomerular, peritubular renal cortical interstitial cells, hepatocytes and liver sinusoidal cells and perimysium, endomysium and nucleoi of skeletal muscle were irisin positive, but its synthesis decreased 1-4h after MI. At all time-points, irisin increased near myocardial connective tissue, with production in skeletal muscle, liver and kidney recovering after 6h, although slower than controls. Unique insight into the pathogenesis of MI is shown, and the gradually decrease of serum irisin might be a diagnostic marker for MI.
Collapse
Affiliation(s)
- Tuncay Kuloglu
- Firat University, School of Medicine, Department of Histology and Embryology, Elazig 23119, Turkey
| | - Suna Aydin
- Department of Cardiovascular Surgery, Elazig Research and Education Hospital, Elazig 23100, Turkey; Firat University, School of Medicine, Department of Anatomy, Elazig 23119, Turkey
| | - Mehmet Nesimi Eren
- Dicle University, School of Medicine, Department of Cardiovascular Surgery, Diyarbakir 21280, Turkey
| | - Musa Yilmaz
- Firat University, School of Medicine, Department of Medical Biochemistry (Firat Hormone Research Groups), Elazig 23119, Turkey
| | - Ibrahim Sahin
- Firat University, School of Medicine, Department of Medical Biochemistry (Firat Hormone Research Groups), Elazig 23119, Turkey; Erzincan University, School of Medicine, Department of Histology and Embryology, Erzincan 24030, Turkey
| | - Mehmet Kalayci
- Firat University, School of Medicine, Department of Medical Biochemistry (Firat Hormone Research Groups), Elazig 23119, Turkey
| | - Emine Sarman
- Firat University, School of Medicine, Department of Histology and Embryology, Elazig 23119, Turkey
| | - Nalan Kaya
- Firat University, School of Medicine, Department of Histology and Embryology, Elazig 23119, Turkey
| | - Osman Fatih Yilmaz
- Firat University, School of Medicine, Department of Histology and Embryology, Elazig 23119, Turkey
| | - Ahmet Turk
- Firat University, School of Medicine, Department of Histology and Embryology, Elazig 23119, Turkey
| | - Yalcin Aydin
- Ankara University, Faculty of Veterinary Medicine, Veterinary Medicine Student, Ankara 06110, Turkey
| | - Mehmet Hanifi Yalcin
- Firat University, Faculty of Veterinary Medicine, Department of Histology and Embryology, Elazig 23119, Turkey
| | - Nimet Uras
- Firat University, School of Medicine, Medical School Student, Elazig 23119, Turkey
| | - Ali Gurel
- Firat University, School of Medicine, Department of Internal Medicine, Elazig 23119, Turkey
| | - Selcuk Ilhan
- Firat University, School of Medicine, Department of Medical Pharmacy, Elazig 23119, Turkey
| | - Evrim Gul
- Department of Emergency, Elazig education and Research Hospital, Elazig 23100, Turkey
| | - Suleyman Aydin
- Firat University, School of Medicine, Department of Medical Biochemistry (Firat Hormone Research Groups), Elazig 23119, Turkey.
| |
Collapse
|
166
|
Bornfeldt KE. 2013 Russell Ross memorial lecture in vascular biology: cellular and molecular mechanisms of diabetes mellitus-accelerated atherosclerosis. Arterioscler Thromb Vasc Biol 2014; 34:705-14. [PMID: 24665124 PMCID: PMC3967130 DOI: 10.1161/atvbaha.113.301928] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 11/22/2013] [Indexed: 01/12/2023]
Abstract
Adults with diabetes mellitus are much more likely to have cardiovascular disease than those without diabetes mellitus. Genetically engineered mouse models have started to provide important insight into the mechanisms whereby diabetes mellitus promotes atherosclerosis. Such models have demonstrated that diabetes mellitus promotes formation of atherosclerotic lesions, progression of lesions into advanced hemorrhaged lesions, and that it prevents lesion regression. The proatherosclerotic effects of diabetes mellitus are driven in part by the altered function of myeloid cells. The protein S100A9 and the receptor for advanced glycation end-products are important modulators of the effect of diabetes mellitus on myelopoiesis, which might promote monocyte accumulation in lesions. Furthermore, myeloid cell expression of the enzyme acyl-CoA synthetase 1 (ACSL1), which converts long-chain fatty acids into their acyl-CoA derivatives, has emerged as causal to diabetes mellitus-induced lesion initiation. The protective effects of myeloid ACSL1-deficiency in diabetic mice, but not in nondiabetic mice, indicate that myeloid cells are activated by diabetes mellitus through mechanisms that play minor roles in the absence of diabetes mellitus. The roles of reactive oxygen species and insulin resistance in diabetes mellitus-accelerated atherosclerosis are also discussed, primarily in relation to endothelial cells. Translational studies addressing whether the mechanisms identified in mouse models are equally important in humans with diabetes mellitus will be paramount.
Collapse
Affiliation(s)
- Karin E Bornfeldt
- From the Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, and Department of Pathology, Diabetes and Obesity Center of Excellence, University of Washington, Seattle, WA
| |
Collapse
|
167
|
Bartova J, Sommerova P, Lyuya-Mi Y, Mysak J, Prochazkova J, Duskova J, Janatova T, Podzimek S. Periodontitis as a risk factor of atherosclerosis. J Immunol Res 2014; 2014:636893. [PMID: 24741613 PMCID: PMC3987959 DOI: 10.1155/2014/636893] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 01/29/2014] [Accepted: 02/17/2014] [Indexed: 11/24/2022] Open
Abstract
Over the last two decades, the amount of evidence corroborating an association between dental plaque bacteria and coronary diseases that develop as a result of atherosclerosis has increased. These findings have brought a new aspect to the etiology of the disease. There are several mechanisms by which dental plaque bacteria may initiate or worsen atherosclerotic processes: activation of innate immunity, bacteremia related to dental treatment, and direct involvement of mediators activated by dental plaque and involvement of cytokines and heat shock proteins from dental plaque bacteria. There are common predisposing factors which influence both periodontitis and atherosclerosis. Both diseases can be initiated in early childhood, although the first symptoms may not appear until adulthood. The formation of lipid stripes has been reported in 10-year-old children and the increased prevalence of obesity in children and adolescents is a risk factor contributing to lipid stripes development. Endothelium damage caused by the formation of lipid stripes in early childhood may lead to bacteria penetrating into blood circulation after oral cavity procedures for children as well as for patients with aggressive and chronic periodontitis.
Collapse
Affiliation(s)
- Jirina Bartova
- Institute of Clinical and Experimental Dental Medicine, First Faculty of Medicine and General University Hospital, Charles University, Karlovo Namesti 32, 12000 Prague, Czech Republic
| | - Pavla Sommerova
- Institute of Clinical and Experimental Dental Medicine, First Faculty of Medicine and General University Hospital, Charles University, Karlovo Namesti 32, 12000 Prague, Czech Republic
| | - Yelena Lyuya-Mi
- Institute of Clinical and Experimental Dental Medicine, First Faculty of Medicine and General University Hospital, Charles University, Karlovo Namesti 32, 12000 Prague, Czech Republic
| | - Jaroslav Mysak
- Institute of Clinical and Experimental Dental Medicine, First Faculty of Medicine and General University Hospital, Charles University, Karlovo Namesti 32, 12000 Prague, Czech Republic
| | - Jarmila Prochazkova
- Institute of Clinical and Experimental Dental Medicine, First Faculty of Medicine and General University Hospital, Charles University, Karlovo Namesti 32, 12000 Prague, Czech Republic
| | - Jana Duskova
- Institute of Clinical and Experimental Dental Medicine, First Faculty of Medicine and General University Hospital, Charles University, Karlovo Namesti 32, 12000 Prague, Czech Republic
| | - Tatjana Janatova
- Institute of Clinical and Experimental Dental Medicine, First Faculty of Medicine and General University Hospital, Charles University, Karlovo Namesti 32, 12000 Prague, Czech Republic
| | - Stepan Podzimek
- Institute of Clinical and Experimental Dental Medicine, First Faculty of Medicine and General University Hospital, Charles University, Karlovo Namesti 32, 12000 Prague, Czech Republic
| |
Collapse
|
168
|
Favero G, Paganelli C, Buffoli B, Rodella LF, Rezzani R. Endothelium and its alterations in cardiovascular diseases: life style intervention. BIOMED RESEARCH INTERNATIONAL 2014; 2014:801896. [PMID: 24719887 PMCID: PMC3955677 DOI: 10.1155/2014/801896] [Citation(s) in RCA: 164] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 01/11/2014] [Indexed: 01/07/2023]
Abstract
The endothelium, which forms the inner cellular lining of blood vessels and lymphatics, is a highly metabolically active organ that is involved in many physiopathological processes, including the control of vasomotor tone, barrier function, leukocyte adhesion, and trafficking and inflammation. In this review, we summarized and described the following: (i) endothelial cell function in physiological conditions and (ii) endothelial cell activation and dysfunction in the main cardiovascular diseases (such as atherosclerosis, and hypertension) and to diabetes, cigarette smoking, and aging physiological process. Finally, we presented the currently available evidence that supports the beneficial effects of physical activity and various dietary compounds on endothelial functions.
Collapse
Affiliation(s)
- Gaia Favero
- Section of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Corrado Paganelli
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Barbara Buffoli
- Section of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Luigi Fabrizio Rodella
- Section of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Rita Rezzani
- Section of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| |
Collapse
|
169
|
Abstract
PURPOSE OF REVIEW Cardiovascular disease (CVD) is the leading cause of morbidity and premature mortality in Europe and the United States, and is increasingly common in developing countries. High-density lipoprotein cholesterol (HDL-C) is an independent risk factor for CVD and is superior to low-density lipoprotein cholesterol (LDL-C) as a predictor of cardiovascular events. The residual risk conferred by low HDL-C in patients with a satisfactory LDL-C was recently highlighted by the European Atherosclerosis Society. Despite the lack of randomized controlled trials, it has been suggested that raising the level of HDL-C should be considered as a therapeutic strategy in high-risk patients because of the strong epidemiological evidence, compelling biological plausibility, and both experimental and clinical research supporting its cardioprotective effects. RECENT FINDINGS Three recent large randomized clinical trials investigating the effect of HDL-C raising with niacin and dalcetrapib in statin-treated patients failed to demonstrate an improvement in cardiovascular outcomes. SUMMARY There is evidence to support the view that HDL functionality and the mechanism by which a therapeutic agent raises HDL-C are more important than plasma HDL-C levels. Future therapeutic agents will be required to improve this functionality rather than simply raising the cholesterol cargo.
Collapse
|
170
|
Favero G, Rodella LF, Reiter RJ, Rezzani R. Melatonin and its atheroprotective effects: a review. Mol Cell Endocrinol 2014; 382:926-37. [PMID: 24291636 DOI: 10.1016/j.mce.2013.11.016] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 11/11/2013] [Accepted: 11/21/2013] [Indexed: 01/20/2023]
Abstract
Atherosclerosis is a chronic vascular disease in which oxidative stress and inflammation are commonly implicated as major causative factors. Identification of novel strategies that contribute to plaque stabilization or inhibition represents a continuing challenge for the medical community. The evidence from the last decade highlights that melatonin influences the cardiovascular system, but its mechanisms of action have not been definitively clarified. Melatonin has atheroprotective effects by acting on different pathogenic signaling processes; these result from its direct free radical scavenger activity, its indirect antioxidant properties and its anti-inflammatory actions. In this review, we summarize the many pieces of the puzzle which identified molecular targets for prevention and therapy against the atherosclerotic pathogenic processes and we evaluate the data documenting that melatonin treatment has important actions that protect against atherosclerosis and atherosclerosis-related cardiovascular diseases.
Collapse
Affiliation(s)
- Gaia Favero
- Section of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Luigi Fabrizio Rodella
- Section of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Russel J Reiter
- Department of Cellular and Structural Biology, UT Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, USA
| | - Rita Rezzani
- Section of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
| |
Collapse
|
171
|
Buja LM. Nikolai N. Anitschkow and the lipid hypothesis of atherosclerosis. Cardiovasc Pathol 2014; 23:183-4. [PMID: 24484612 DOI: 10.1016/j.carpath.2013.12.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 12/23/2013] [Indexed: 11/25/2022] Open
Abstract
One hundred years ago, Nikolai N. Anitschkow published his seminal observations that rabbits fed a diet with only high-cholesterol developed atheromatous lesions in association with greatly elevated blood cholesterol. For many years, Anitschkow's observations received little recognition. However, eventually a combination of experimental work, autopsy studies, epidemiological investigation and clinical trials has led to the firm establishment of the essential role of lipids in the response to injury theory of atherosclerosis. The twists and turns in the acceptance of the lipid hypothesis of atherosclerosis is briefly reviewed. Today, it is well worth celebrating the 100th anniversary of the seminal insight into human vascular disease of the brilliant experimental pathologist, Nikolai N. Anitschkow.
Collapse
Affiliation(s)
- L Maximilian Buja
- Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, TX, USA.
| |
Collapse
|
172
|
Chattopadhyay R, Dyukova E, Singh NK, Ohba M, Mobley JA, Rao GN. Vascular endothelial tight junctions and barrier function are disrupted by 15(S)-hydroxyeicosatetraenoic acid partly via protein kinase C ε-mediated zona occludens-1 phosphorylation at threonine 770/772. J Biol Chem 2013; 289:3148-63. [PMID: 24338688 DOI: 10.1074/jbc.m113.528190] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Disruption of tight junctions (TJs) perturbs endothelial barrier function and promotes inflammation. Previously, we have shown that 15(S)-hydroxyeicosatetraenoic acid (15(S)-HETE), the major 15-lipoxygenase 1 (15-LO1) metabolite of arachidonic acid, by stimulating zona occludens (ZO)-2 tyrosine phosphorylation and its dissociation from claudins 1/5, induces endothelial TJ disruption and its barrier dysfunction. Here, we have studied the role of serine/threonine phosphorylation of TJ proteins in 15(S)-HETE-induced endothelial TJ disruption and its barrier dysfunction. We found that 15(S)-HETE enhances ZO-1 phosphorylation at Thr-770/772 residues via PKCε-mediated MEK1-ERK1/2 activation, causing ZO-1 dissociation from occludin, disrupting endothelial TJs and its barrier function, and promoting monocyte transmigration; these effects were reversed by T770A/T772A mutations. In the arteries of WT mice ex vivo, 15(S)-HETE also induced ZO-1 phosphorylation and endothelial TJ disruption in a PKCε and MEK1-ERK1/2-dependent manner. In line with these observations, in WT mice high fat diet feeding induced 12/15-lipoxygenase (12/15-LO) expression in the endothelium and caused disruption of its TJs and barrier function. However, in 12/15-LO(-/-) mice, high fat diet feeding did not cause disruption of endothelial TJs and barrier function. These observations suggest that the 12/15-LO-12/15(S)-HETE axis, in addition to tyrosine phosphorylation of ZO-2, also stimulates threonine phosphorylation of ZO-1 in the mediation of endothelial TJ disruption and its barrier dysfunction.
Collapse
Affiliation(s)
- Rima Chattopadhyay
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | | | | | | | | | | |
Collapse
|
173
|
Mulas MF, Maxia A, Dessì S, Mandas A. Cholesterol esterification as a mediator of proliferation of vascular smooth muscle cells and peripheral blood mononuclear cells during atherogenesis. J Vasc Res 2013; 51:14-26. [PMID: 24280911 DOI: 10.1159/000355218] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 08/19/2013] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND/AIMS We determined growth rates, cholesterol esterification and mRNA levels for caveolin-1 (Cav-1), neutral cholesterol esters hydrolase (n-CEH) and ATP-binding cassette transporter (ABCA-1), in quiescent and growth-stimulated peripheral blood mononuclear cells (PBMCs) and intimal vascular smooth muscle cells (VSMCs) from blood and primary atherosclerotic plaques, respectively. These cells were cultured in the presence or absence of the mTOR inhibitor 40-O-(2-hydroxyethyl) rapamycin (RAD). METHODS The rate of cell proliferation was determined by 3H-thymidine incorporation into DNA and that of lipid metabolism by utilizing 14C-acetate and 14C-oleate as precursors. Lipid deposit in the vascular cells was evaluated by Oil Red O staining and lipid mass by thin layer chromatography-linked enzymatic assay. RESULTS Growth stimulation of PBMCs and VSMCs caused a rapid increase in intracellular cholesterol esterification and an accumulation of cholesterol esters (CEs) accompanied by a reduction of free cholesterol (FC) and Cav-1, ABCA-1 and n-CEH mRNAs. RAD reduced intracellular lipid accumulation in growth-stimulated cells and also increased expression of Cav-1, n-CEH and ABCA-1 genes. CONCLUSION Collectively, these data provide evidence that the determination of CEs in PBMCs may be an easy prescreening test to identify subjects at risk for vascular proliferative disease and that FC, CE, Cav-1, n-CEH and ABCA-1 may be suitable targets for antiproliferative therapies.
Collapse
Affiliation(s)
- Maria Franca Mulas
- Dipartimento di Scienze Mediche, University of Cagliari, Monserrato, Italy
| | | | | | | |
Collapse
|
174
|
Wu J, Chen S, Liu L, Gao X, Zhou Y, Wang C, Zhang Q, Wang A, Hussain M, Sun B, Wu S, Zhao X. Non-high-density lipoprotein cholesterolvslow-density lipoprotein cholesterol as a risk factor for ischemic stroke: a result from the Kailuan study. Neurol Res 2013; 35:505-11. [PMID: 23594748 DOI: 10.1179/1743132813y.0000000206] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Affiliation(s)
- Jianwei Wu
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shengyun Chen
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Liping Liu
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiang Gao
- Department of NutritionHarvard University School of Public Health, Boston, MA, USA
| | - Yong Zhou
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Chunxue Wang
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qian Zhang
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Cell Transplantationthe General Hospital of Chinese People's Armed Police Forces, Beijing, China
| | - Anxin Wang
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical University, Beijing, China
| | | | - Baoying Sun
- Department of NeurologyJinan Central Hospital, Shandong, China
| | - Shouling Wu
- Department of CardiologyKailuan Hospital, Tangshan, China
| | - Xingquan Zhao
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
175
|
Xiao Q, Zhang F, Grassia G, Hu Y, Zhang Z, Xing Q, Yin X, Maddaluno M, Drung B, Schmidt B, Maffia P, Ialenti A, Mayr M, Xu Q, Ye S. Matrix metalloproteinase-8 promotes vascular smooth muscle cell proliferation and neointima formation. Arterioscler Thromb Vasc Biol 2013; 34:90-8. [PMID: 24158518 DOI: 10.1161/atvbaha.113.301418] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE We investigated the role of matrix metalloproteinase-8 (MMP8) in neointima formation and in vascular smooth muscle cell (VSMC) migration and proliferation. APPROACH AND RESULTS After carotid artery wire injuring, MMP8(-/-)/apoE(-/-) mice had fewer proliferating cells in neointimal lesions and smaller lesion sizes. Ex vivo assays comparing VSMCs isolated from MMP8 knockout and wild-type mice showed that MMP8 knockout decreased proliferation and migration. Proteomics analysis revealed that a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) had lower concentrations in MMP8 knockout VSMC culture media than in MMP8 wild-type VSMC culture media. Western blot, flow cytometric, and immunocytochemical analyses showed that MMP8 knockout VSMCs contained more pro-ADAM10 but less mature ADAM10, more N-cadherin, and β-catenin in the plasma membrane but less β-catenin in the nucleus and less cyclin D1. Treatment of MMP8 wild-type VSMCs with an ADAM10 inhibitor, GI254023X, or siRNA knockdown of ADAM10 in MMP8 wild-type VSMCs inhibited proliferation and migration, increased N-cadherin and β-catenin in the plasma membrane, reduced β-catenin in the nucleus, and decreased cyclin D1 expression. Incubation of MMP8 knockout VSMCs with a recombinant ADAM10 rescued the proliferative and migratory ability of MMP8 knockout VSMCs and increased cyclin D1 expression. Furthermore, immunohistochemical analyses showed colocalization of ADAM10 with VSMCs and N-cadherin, and nuclear accumulation of β-catenin in the neointima in apoE(-/-)/MMP8(+/+) mice. CONCLUSIONS MMP8 enhances VSMC proliferation via an ADAM10, N-cadherin, and β-catenin-mediated pathway and plays an important role in neointima formation.
Collapse
Affiliation(s)
- Qingzhong Xiao
- From William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom (Q. Xiao, F.Z., S.Y.); Department of Cardiology, Peking University People's Hospital, Beijing, China (F.Z.); Department of Pharmacy, University of Naples Federico II, Naples, Italy (G.G., M. Maddaluno, P.M., A.I.); Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Y.H., Z.Z., Q. Xing, X.Y., M. Mayr, Q. Xu); Clemens Schöpf Institute, Technische Universität Darmstadt, Darmstadt, Germany (B.D., B.S.); Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (P.M.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
176
|
Li J, Zhang K, Yang P, Liao Y, Wu L, Chen J, Zhao A, Li G, Huang N. Research of smooth muscle cells response to fluid flow shear stress by hyaluronic acid micro-pattern on a titanium surface. Exp Cell Res 2013; 319:2663-72. [DOI: 10.1016/j.yexcr.2013.05.027] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 05/19/2013] [Accepted: 05/29/2013] [Indexed: 12/13/2022]
|
177
|
Gilbert RE. Augmenting Endothelial Repair in Diabetes: Role of Bone Marrow-Derived Cells. Can J Diabetes 2013; 37:315-8. [DOI: 10.1016/j.jcjd.2013.06.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 06/23/2013] [Accepted: 06/24/2013] [Indexed: 01/09/2023]
|
178
|
Bi Y, Zhong H, Xu K, Qi X. Combination of Periaortic Elastase Incubation and Cholesterol-Rich Diet: A Novel Model of Atherosclerosis in Rabbit Abdominal Aorta. Cell Biochem Biophys 2013; 68:611-4. [DOI: 10.1007/s12013-013-9753-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
179
|
Bhardwaj P, Khanna D. Green tea catechins: defensive role in cardiovascular disorders. Chin J Nat Med 2013; 11:345-53. [DOI: 10.1016/s1875-5364(13)60051-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Indexed: 10/26/2022]
|
180
|
Abstract
Functional integrity of endothelial cells is an indicator and a prerequisite for vascular health and counteracts the development of atherosclerosis. This concept of 'endothelial therapy' was developed in the late 1990s as an approach to preserve or restore endothelial cell health given that 'the knowledge of the mechanisms involved in 'endothelial dysfunction' allows us to interfere specifically with pathogenic pathways at very early time points and to slow down the progression of disease'. In the present review, the principles underlying endothelial cell health will be discussed as well as the role of endothelial therapy as a preventive measure to reduce the prevalence of coronary artery disease or to delay disease progression in patients with chronic coronary artery disease. This article also highlights the importance of active participation, the need to reduce the number of future patients in view of the rising prevalence of childhood obesity, and the potential of endothelial therapy to improve survival, reduce disability and health costs, and to improve overall quality of life in patients at risk for or already diagnosed with coronary artery disease. The preventive and therapeutic approaches and considerations described herein can be applied by physicians, patients, parents, educators, health agencies, and political decision makers to help reducing the global cardiovascular disease burden in the decades to come.
Collapse
Affiliation(s)
- Matthias Barton
- Molecular Internal Medicine, University of Zürich, LTK Y44 G22, Winterthurerstrasse 190, 8057 Zürich, Switzerland.
| |
Collapse
|
181
|
Hidalgo A, Tello L, Toro EF. Numerical and analytical study of an atherosclerosis inflammatory disease model. J Math Biol 2013; 68:1785-814. [PMID: 23719743 DOI: 10.1007/s00285-013-0688-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 05/06/2013] [Indexed: 11/24/2022]
Abstract
We study a reaction-diffusion mathematical model for the evolution of atherosclerosis as an inflammation process by combining analytical tools with computer-intensive numerical calculations. The computational work involved the calculation of more than sixty thousand solutions of the full reaction-diffusion system and lead to the complete characterisation of the ω-limit for every initial condition. Qualitative properties of the solution are rigorously proved, some of them hinted at by the numerical study.
Collapse
Affiliation(s)
- A Hidalgo
- Dept. Matemática Aplicada y Métodos Informáticos. E.T.S.I., Minas, Universidad Politécnica de Madrid, Rios Rosas 21, 28003 , Madrid, Spain,
| | | | | |
Collapse
|
182
|
Biodegradable synthetic high-density lipoprotein nanoparticles for atherosclerosis. Proc Natl Acad Sci U S A 2013; 110:9445-50. [PMID: 23671083 DOI: 10.1073/pnas.1301929110] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Atherosclerosis remains one of the most common causes of death in the United States and throughout the world because of the lack of early detection. Macrophage apoptosis is a major contributor to the instability of atherosclerotic lesions. Development of an apoptosis targeted high-density lipoprotein (HDL)-mimicking nanoparticle (NP) to carry contrast agents for early detection of vulnerable plaques and the initiation of preventative therapies that exploit the vascular protective effects of HDL can be attractive for atherosclerosis. Here, we report the construction of a synthetic, biodegradable HDL-NP platform for detection of vulnerable plaques by targeting the collapse of mitochondrial membrane potential that occurs during apoptosis. This HDL mimic contains a core of biodegradable poly(lactic-co-glycolic acid), cholesteryl oleate, and a phospholipid bilayer coat that is decorated with triphenylphosphonium (TPP) cations for detection of mitochondrial membrane potential collapse. The lipid layer provides the surface for adsorption of apolipoprotein (apo) A-I mimetic 4F peptide, and the core contains diagnostically active quantum dots (QDs) for optical imaging. In vitro uptake, detection of apoptosis, and cholesterol binding studies indicated promising detection ability and therapeutic potential of TPP-HDL-apoA-I-QD NPs. In vitro studies indicated the potential of these NPs in reverse cholesterol transport. In vivo biodistribution and pharmacokinetics indicated favorable tissue distribution, controlled pharmacokinetic parameters, and significant triglyceride reduction for i.v.-injected TPP-HDL-apoA-I-QD NPs in rats. These HDL NPs demonstrate excellent biocompatibility, stability, nontoxic, and nonimmunogenic properties, which prove to be promising for future translation in early plaque diagnosis and might find applications to prevent vulnerable plaque progression.
Collapse
|
183
|
Kundumani-Sridharan V, Dyukova E, Hansen DE, Rao GN. 12/15-Lipoxygenase mediates high-fat diet-induced endothelial tight junction disruption and monocyte transmigration: a new role for 15(S)-hydroxyeicosatetraenoic acid in endothelial cell dysfunction. J Biol Chem 2013; 288:15830-42. [PMID: 23589307 DOI: 10.1074/jbc.m113.453290] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
A convincing body of evidence suggests that 12/15-lipoxygenase (12/15-LO) plays a role in atherosclerosis. However, the mechanisms of its involvement in the pathogenesis of this disease are not clear. Therefore, the purpose of this study is to understand the mechanisms by which 12/15-LO mediates endothelial dysfunction. 15(S)-Hydroxyeicosatetraenoic acid (15(S)-HETE), the major 12/15-LO metabolite of arachidonic acid (AA), induced endothelial barrier permeability via Src and Pyk2-dependent zonula occluden (ZO)-2 tyrosine phosphorylation and its dissociation from the tight junction complexes. 15(S)-HETE also stimulated macrophage adhesion to the endothelial monolayer in Src and Pyk2-dependent manner. Ex vivo studies revealed that exposure of arteries from WT mice to AA or 15(S)-HETE led to Src-Pyk2-dependent ZO-2 tyrosine phosphorylation, tight junction disruption, and macrophage adhesion, whereas the arteries from 12/15-LO knock-out mice are protected from these effects of AA. Feeding WT mice with a high-fat diet induced the expression of 12/15-LO in the arteries leading to tight junction disruption and macrophage adhesion and deletion of the 12/15-LO gene disallowed these effects. Thus, the findings of this study provide the first evidence of the role of 12/15-LO and its AA metabolite, 15(S)-HETE, in high-fat diet-induced endothelial tight junction disruption and macrophage adhesion, the crucial events underlying the pathogenesis of atherosclerosis.
Collapse
|
184
|
Abstract
Tissue factor (TF) is abundantly present in atherosclerotic plaques and it is the primary source of TF that triggers the rapid activation of the coagulation cascade after plaque rupture. While much of this TF is associated with monocyte/macrophages and vascular smooth muscle cells, recent studies suggests TF-positive microparticles (MPs) are the most abundant source in plaques. Further, while intravascular TF is largely absent in healthy patients, cardiovascular disease patients have increased TF expression in circulating monocytes, which can result in increased levels of TF-positive MPs. This brief review describes how TF is the primary initiator of atherothrombosis and how TF-positive MPs may serve as a biomarker to identify patients at greater risk of forming an occlusive thrombus. In addition, currently used therapeutics, such as statins and inhibitors of the renin angiotensin system, may have additional benefits by reducing TF expression and subsequent thrombosis.
Collapse
Affiliation(s)
- A Phillip Owens
- Department of Medicine, Division of Hematology and Oncology, McAllister Heart Institute, University of North Carolina at Chapel Hill, 98 Manning Drive Campus Box 7035, Chapel Hill, NC 27599, USA.
| | | |
Collapse
|
185
|
Abstract
Atherosclerosis underlies coronary artery disease (CAD) and cerebrovascular disease, which are the most common forms of life-threatening cardiovascular disorders. To minimize the risk of atherosclerotic complications, primary and secondary prevention strategies seek to control risk factors. Reducing low-density lipoprotein (LDL) cholesterol through lipid-lowering drugs, such as statins, in particular yields a proportional decrease in cardiovascular disease risk. Atherosclerosis is considered to be a complex chronic inflammatory process triggered by cardiovascular risk factors which cause endothelial dysfunction and inflammatory cell infiltration within the artery wall. In this review, we summarize the current understanding of the underling molecular mechanisms of the immune signals in the development and progression of atherosclerosis. Among various molecular mechanisms, toll like receptors (TLRs) are potent proinflammatory cytokines that operate to induce inflammation play an important role in the pathogenesis of atherosclerosis. Moreover, we discuss current knowledge regarding monocyte/macrophage biology that contributes to the progression of atherosclerosis, including macrophage polarization and heterogeneity. Understanding the molecular mechanisms in conjunction with orchestration of monocyte/macrophage biology should provide a basis for novel treatment strategies to prevent the development and progression of atherosclerosis.
Collapse
Affiliation(s)
- Hiroshi Iwata
- Brigham and Women's Hospital, Department of Medicine, Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, 3 Blackfan Street, 17th Floor, Boston, MA 02115, USA.
| | | |
Collapse
|
186
|
Ouimet M. Autophagy in obesity and atherosclerosis: Interrelationships between cholesterol homeostasis, lipoprotein metabolism and autophagy in macrophages and other systems. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:1124-33. [PMID: 23545567 DOI: 10.1016/j.bbalip.2013.03.007] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 03/18/2013] [Accepted: 03/20/2013] [Indexed: 12/14/2022]
Abstract
The incidence of diseases characterized by a dysregulation of lipid metabolism such as obesity, diabetes and atherosclerosis is rising at alarming rates, driving research to uncover new therapies to manage dyslipidemias and resolve the metabolic syndrome conundrum. Autophagy and lipid homeostasis - both ancient cellular pathways - have seemingly co-evolved to share common regulatory elements, and autophagy has emerged as a prominent mechanism involved in the regulation of lipid metabolism. This review highlights recent findings on the role of autophagy in the regulation of cellular cholesterol homeostasis and lipoprotein metabolism, with special emphasis on macrophages. From modulation of inflammation to regulation of cellular cholesterol levels, a protective role for autophagy in atherosclerosis is emerging. The manipulation of autophagic activity represents a new possible therapeutic approach for the treatment complex metabolic disorders such as obesity and the metabolic syndrome.
Collapse
Affiliation(s)
- Mireille Ouimet
- Department of Medicine, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
187
|
Sano E, Tashiro S, Tadakuma H, Takei T, Ueda T, Tsumoto K. Type 1 IFN inhibits the growth factor deprived apoptosis of cultured human aortic endothelial cells and protects the cells from chemically induced oxidative cytotoxicity. J Cell Biochem 2013; 113:3823-34. [PMID: 22821369 DOI: 10.1002/jcb.24259] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
It has been shown that the genesis of atherosclerotic lesions is resulted from the injury of vascular endothelial cells and the cell damage is triggered by oxygen radicals generated from various tissues. Human vascular endothelial cells can survive and proliferate depending on growth factors such as VEGF or basic FGF and are induced apoptosis by the deprivation of growth factor or serum. It was found that type 1 IFN inhibits the growth factor deprived cell death of human aortic endothelial cells (HAEC) and protects the cells from chemically induced oxidative cytotoxicity. The anti-apoptotic effects of type 1 IFN were certified by flow cytometry using annexin-V-FITC/PI double staining and cell cycle analysis, fluorescence microscopy using Hoechst33342 and PI, colorimetric assay for caspase-3 activity, p53 and bax mRNA expressions, and cell counts. It was considered that IFN-β inhibits the executive late stage apoptosis from the results of annexin-V-FITC/PI double staining and the inhibition of caspase-3 activity, and that the anti-apoptotic effect might be owing to the direct inhibition of the apoptotic pathway mediated by p53 from the transient down-regulation of bax mRNA expression. Whereas, type 1 IFN protected the cells from the oxidative cytotoxicity induced by tertiary butylhydroperoxide (TBH) under the presence of Ca(2+). The effects of IFN-β is more potent inhibitor of cell death than IFN-α. These results indicate that type 1 IFN, especially IFN-β may be useful for the diseases with vascular endothelium damage such as atherosclerosis or restenosis after angioplasty as a medical treatment or a prophylactic.
Collapse
Affiliation(s)
- Emiko Sano
- Department of Medical Proteomics Laboratory, Institute of Medical Science, The University of Tokyo, Shirokanedai, Tokyo, 108-8639, Japan.
| | | | | | | | | | | |
Collapse
|
188
|
Ozaki MR, de Almeida EA. Evolution and involution of atherosclerosis and its relationship with vascular reactivity in hypercholesterolemic rabbits. ACTA ACUST UNITED AC 2013; 65:297-304. [DOI: 10.1016/j.etp.2011.09.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Revised: 07/24/2011] [Accepted: 09/25/2011] [Indexed: 10/16/2022]
|
189
|
Wei Y, Nazari-Jahantigh M, Neth P, Weber C, Schober A. MicroRNA-126, -145, and -155: a therapeutic triad in atherosclerosis? Arterioscler Thromb Vasc Biol 2013; 33:449-54. [PMID: 23324496 DOI: 10.1161/atvbaha.112.300279] [Citation(s) in RCA: 168] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Atherosclerosis is a condition caused by lipid-induced inflammation of the vessel wall orchestrated by a complex interplay of various cell types, such as endothelial cells, smooth muscle cells, and macrophages. MicroRNAs (miRNAs) have emerged as key regulators of gene expression typically by repressing the target mRNA, which determines cell fate and function under homeostatic and disease conditions. Here, we outline the effects of miRNA-145, -126, and -155 in atherosclerosis in vivo. Downregulation of miR-145, which controls differentiation of smooth muscle cells, promotes lesion formation, whereas the endothelial cell-specific miRNA-126 signals the need for endothelial repair through its transfer from apoptotic endothelial cells in microvesicles. Elevated miR-155 levels are characteristic of proinflammatory macrophages and atherosclerotic lesions. However, the effects of miR-155 seem to be different in early and advanced atherosclerosis. The discovery of the role of these miRNAs in atherosclerosis sheds light on the current concepts of atherogenesis and may provide novel treatment options for cardiovascular diseases.
Collapse
Affiliation(s)
- Yuanyuan Wei
- Experimental Vascular Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | | | | | | | | |
Collapse
|
190
|
Przybylowski P, Koc-Zorawska E, Malyszko J, Mysliwiec M, Malyszko J. Renalase and Endothelial Dysfunction in Heart Transplant Recipients. Transplant Proc 2013; 45:394-6. [DOI: 10.1016/j.transproceed.2012.02.042] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 02/28/2012] [Indexed: 12/21/2022]
|
191
|
Abstract
Despite advances in prevention and treatment, atherosclerotic vascular disease continues to account for significant morbidity, mortality, and economic burden in the western world. Our current understanding of this disease presents atherosclerosis as a chronic inflammatory process involving multiple cell types in various stages of activation, apoptosis, and necrosis. These cells include monocyte/macrophage, dendritic cells, lymphocytes, endothelial cells, and vascular smooth muscle cells. Activation of these cells and their processes is initiated and sustained by a complex network of soluble factors termed cytokines. Cytokines are produced and recognized by both inflammatory and resident vascular cells, allowing crosstalk between these two systems. Cytokines also regulate the phenotype of many of these cell types. Recognizing functions of these cytokines and their effects on cells which populate atherosclerotic plaque is key to uncovering targets of therapeutic intervention. This paper will present recent studies which describe the cellular protagonists of atherosclerosis and the role they play in formation of atherosclerotic plaque. It will also describe the cytokines which have been identified as produced by and directly affecting dysfunction of these cells. Because atherosclerosis is considered an inflammatory condition, emphasis will be placed on inflammatory cytokines and their effects on atherogenesis. We will conclude with new directions in therapeutic strategies and points of emphasis for future research.
Collapse
Affiliation(s)
- Michael V. Autieri
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center and Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Room 1050, MERB, 3500 North Broad Street, Philadelphia, PA 19140, USA
| |
Collapse
|
192
|
Pandey R, Gupta S, Lal H, Mehta HC, Aggarwal SK. Hyperhomocysteinemia and cardiovascular disease: The nutritional perspectives. Indian J Clin Biochem 2012; 15:20-30. [PMID: 23105265 DOI: 10.1007/bf02867541] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Several members of the vitamin B-complex family are known to participate in the normal metabolism of homocysteine (Hcy). Leaving aside the genetic determinants of hyperhomocysteinemia (HHC), the deficiencies of these vitamins can also result in HHC. The situation of sustained and long standing HHC is likely to be prevalent in population groups with low/average socio-economic status, geriatric population and alcohol abusers. If not corrected by supplementation, these population groups certainly are more vulnerable to develop atherosclerosis (AS) and subsequently, cardiovascular disease (CVD). Hyperhomocysteinemia per se and/or HHC-induced oxidative stress result(s) in chronic chemical endothelial injury/dysfunction, smooth muscle proliferation, prothrombotic state and oxidation of low density lipoproteins (LDL) leading to diverse cardiovascular complications. In the first decade of the new millennium, major research efforts would be directed towards understanding the basic mechanism of HHC-induced oxidative stress and the pathophysiology of HHC-induced CVD, culminating in the evolution of hitherto unknown therapeutic strategies such as nutriceuticals and oxidant-antidotes.
Collapse
Affiliation(s)
- R Pandey
- Department of Biochemistry, Pt. B.D. Sharma, Post Graduate Institute of Medical Sciences, Shanti Sadan, 6-Gole Market, 124001 Model Town, Rohtak (Haryana)
| | | | | | | | | |
Collapse
|
193
|
Nguyen AT, Gomez D, Bell RD, Campbell JH, Clowes AW, Gabbiani G, Giachelli CM, Parmacek MS, Raines EW, Rusch NJ, Speer MY, Sturek M, Thyberg J, Towler DA, Weiser-Evans MC, Yan C, Miano JM, Owens GK. Smooth muscle cell plasticity: fact or fiction? Circ Res 2012; 112:17-22. [PMID: 23093573 DOI: 10.1161/circresaha.112.281048] [Citation(s) in RCA: 133] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Anh T Nguyen
- University of Virginia, Cardiovascular Research Center, Charlottesville, VA 22908, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
194
|
Azahri NSM, Di Bartolo BA, Khachigian LM, Kavurma MM. Sp1, acetylated histone-3 and p300 regulate TRAIL transcription: mechanisms of PDGF-BB-mediated VSMC proliferation and migration. J Cell Biochem 2012; 113:2597-606. [PMID: 22415975 DOI: 10.1002/jcb.24135] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
We recently reported that TNF-related apoptosis-inducing ligand (TRAIL) is important in atherogenesis, since it can induce vascular smooth muscle cell (VSMC) proliferation and arterial thickening following injury. Here we show the first demonstrate that TRAIL siRNA reduces platelet-derived growth factor-BB (PDGF-BB)-stimulated VSMC proliferation and migration. PDGF-BB-inducible VSMC proliferation was completely inhibited in VSMCs isolated from aortas of TRAIL(-/-) mice; whereas inducible migration was blocked compared to control VSMCs. TRAIL transcriptional control mediating this response is not established. TRAIL mRNA, protein and promoter activity was increased by PDGF-BB and subsequently inhibited by dominant-negative Sp1, suggesting that the transcription factor Sp1 plays a role. Sp1 bound multiple Sp1 sites on the TRAIL promoter, including two established (Sp1-1 and -2) and two novel Sp1-5/6 and -7 sites. PDGF-BB-inducible TRAIL promoter activity by Sp1 was mediated through these sites, since transverse mutations to each abolished inducible activity. PDGF-BB stimulation increased acetylation of histone-3 (ac-H3) and expression of the transcriptional co-activator p300, implicating chromatin remodelling. p300 overexpression increased TRAIL promoter activity, which was blocked by dominant-negative Sp1. Furthermore, PDGF-BB treatment increased the physical interaction of Sp1, p300 and ac-H3, while chromatin immunoprecipitation studies revealed Sp1, p300 and ac-H3 enrichment on the TRAIL promoter. Taken together, our studies demonstrate for the first time that PDGF-BB-induced TRAIL transcriptional activity requires the cooperation of Sp1, ac-H3 and p300, mediating increased expression of TRAIL which is important for VSMC proliferation and migration. Our findings have the promising potential for targeting TRAIL as a new therapeutic for vascular proliferative disorders.
Collapse
Affiliation(s)
- Nor Saadah M Azahri
- Centre for Vascular Research, University of New South Wales, Sydney, NSW 2052, Australia
| | | | | | | |
Collapse
|
195
|
Scholtes VPW, de Vries JPPM, Catanzariti LM, de Kleijn DPV, Moll FL, de Borst GJ, Pasterkamp G. Biobanking in atherosclerotic disease, opportunities and pitfalls. Curr Cardiol Rev 2012; 7:9-14. [PMID: 22294969 PMCID: PMC3131717 DOI: 10.2174/157340311795677707] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2010] [Revised: 10/16/2010] [Accepted: 01/07/2011] [Indexed: 01/10/2023] Open
Abstract
Cardiovascular disease is the leading cause of death in Western countries and current research is still focusing on optimizing therapeutic approaches in the battle against this multifactorial disease. Concepts regarding the pathogenesis of many cardiovascular diseases originate from observations of human atherosclerotic tissue obtained from autopsies or during vascular surgery. These observations have helped us to disentangle the pathophysiology of atherosclerosis. However, identifying vulnerable patients, those prone to developing cardiovascular complications, remains difficult. The search for predictive cardiovascular biomarkers continues and large, well organized biobanks are needed to discover or validate novel biomarkers. Biobanks are an extremely valuable resource that enables us to study the influence of both genetic and environmental factors on the development of multifactorial diseases such as atherosclerosis. This review will focus on the advantages and pitfalls in atherosclerotic biobanking.
Collapse
Affiliation(s)
- V P W Scholtes
- Experimental Laboratory Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
196
|
Wong BW, Meredith A, Lin D, McManus BM. The biological role of inflammation in atherosclerosis. Can J Cardiol 2012; 28:631-41. [PMID: 22985787 DOI: 10.1016/j.cjca.2012.06.023] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 06/26/2012] [Accepted: 06/27/2012] [Indexed: 01/11/2023] Open
Abstract
The concept of the involvement of inflammation in the pathogenesis of atherosclerosis has existed since the 1800s, stemming from sentinel pathologic observations made by Rudolf Virchow, Karl Rokitansky, and others. Our understanding of the complex role played by immune and inflammatory mediators in the initiation and progression of atherosclerosis has evolved considerably in the intervening years, and today, a dramatically evolved understanding of these processes has led to advances in both diagnostic and prognostic approaches, as well as novel treatment modalities targeting inflammatory and immune mediators. Therapeutic interventions working through multiple mechanisms involved in atheroma pathogenesis, such as statins, which both lower lipids and alter the inflammatory milieu in the vessel wall, hold promise for the future. In this brief review, we explore the biological role of inflammation in atherosclerosis, with a focus on cellular involvement in both acute and chronic inflammation, and outline novel biomarkers of inflammation and atherosclerosis with a particular focus on the potential application of these novel approaches in improving strategies for disease diagnosis and management.
Collapse
Affiliation(s)
- Brian W Wong
- UBC James Hogg Research Centre, Institute for Heart and Lung Health, St Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| | | | | | | |
Collapse
|
197
|
Huang X, Wang F, Chen W, Li Z, Wang N, Chen Y, von Maltzan K. Dao-Tan decoction inhibits tumor necrosis factor-α-induced intercellular adhesion molecule-1 expression by blocking JNK and p38 signaling pathways in human umbilical vein endothelial cells. PHARMACEUTICAL BIOLOGY 2012; 50:1111-1117. [PMID: 22762513 DOI: 10.3109/13880209.2012.658476] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
CONTEXT Dao-Tan decoction (DTD) is a Chinese herb prescription used to treat atherosclerosis or dizziness for centuries. Previous study shows that DTD could inhibit intercellular adhesion molecule-1 (ICAM-1) expression induced by tumor necrosis factor-α (TNF-α). However, its mechanism has never been clearly described. OBJECTIVE To examine the hypothesis that DTD might inhibit TNF-α-induced ICAM-1 expression through regulating the mitogen-activated protein kinase (MAPK) pathways, involving Jun N-terminal kinase (JNK) and p38. MATERIALS AND METHODS The rats were orally administrated with DTD for 3 days (2.3 g/kg per day), then the serum was collected. Human umbilical vein endothelial cells (HUVECs) were cultured and stimulated by TNF-α with or without DTD serum. The expression of ICAM-1 mRNA was examined by reverse transcription-polymerase chain reaction and the expression of p38 and JNK was examined by Western blot analysis. RESULTS DTD serum significantly inhibits TNF-α-induced ICAM-1 expression by 17-41% on HUVECs. TNF-α-induced JNK and p38 activations, which were involved in ICAM-1 expression, were significantly inhibited with DTD serum treatment by 10-50% on HUVEC. DISCUSSION AND CONCLUSION Based on the theory of traditional Chinese medicine (TCM), pathogenesis of atherosclerosis is caused by "blood" and "phlegm" attached on blood vessels. DTD has a function of "dissolving phlegm", thus it is chosen for the treatment of atherosclerosis. This study demonstrated that DTD could inhibit the expression of ICAM-1, by significantly preventing the activation of JNK and p38, which are important factors of atherosclerosis. Therefore, the present study indicates the pharmacological basis for treatment of atherosclerosis with DTD.
Collapse
Affiliation(s)
- Xiaobo Huang
- Department of Chinese Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | | | | | | | | | | | | |
Collapse
|
198
|
Gu Z, Rolfe BE, Xu ZP, Campbell JH, Lu GQM, Thomas AC. Antibody-targeted drug delivery to injured arteries using layered double hydroxide nanoparticles. Adv Healthc Mater 2012. [PMID: 23184804 DOI: 10.1002/adhm.201200069] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Targeted local delivery of a nanoparticle-based, antibody-targeted, and low molecular weight heparin (LMWH) delivery system successfully reduces restenosis and thrombus formation in an animal model. An antibody recognizing cross-linked fibrin (XLF) D-dimer is successfully conjugated to layered double hydroxide nanoparticles. Use of the anti-XLF-conjugated LMWH-carrying layered double hydroxide nanoparticles shows successful targeting of the nanoparticles (red) to the injured artery wall (green), resulting in decreased neointimal thickening and thrombus formation.
Collapse
Affiliation(s)
- Zi Gu
- ARC Centre of Excellence for Functional Nanomaterials, Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | | | | | | | | | | |
Collapse
|
199
|
Abstract
PURPOSE OF REVIEW HDL cholesterol concentration is inversely correlated with cardiovascular disease and has a wide range of functions involved in many systems. The purpose of this review is to summarize HDL functionality, its relevance to atherosclerosis and factors affecting HDL functions. RECENT FINDINGS The contribution of HDL to reverse cholesterol transport may not be as great as first envisaged. However, it still plays an important role in cholesterol efflux from peripheral tissues. The capacity of HDL to promote cellular cholesterol efflux in an ex-vivo model has been reported to correlate more closely with carotid intima-media thickness than HDL cholesterol concentration. Recently, a variety of other functions of HDL have been described including antimicrobial, antioxidant, antiglycation, anti-inflammatory, nitric oxide--inducing, antithrombotic and antiatherogenic activity and immune modulation as well as a potential role in glucose homeostasis, diabetes pathophysiology and complications. SUMMARY HDL has a wide range of functions some of which are independent of its cholesterol content. Its cargo of apolipoproteins, various proteins and phospholipids contributes most to its various functions. These functions are affected by a number of genetic, physiological and pathological factors.
Collapse
Affiliation(s)
- Handrean Soran
- University Department of Medicine, Central Manchester and Manchester Children University Hospital NHS Foundation Trust, School of Biomedicine, University of Manchester, Manchester, UK
| | | | | | | |
Collapse
|
200
|
Barton M. Position paper: The membrane estrogen receptor GPER--Clues and questions. Steroids 2012; 77:935-42. [PMID: 22521564 DOI: 10.1016/j.steroids.2012.04.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Revised: 03/13/2012] [Accepted: 04/01/2012] [Indexed: 12/25/2022]
Abstract
Rapid signaling of estrogen involves membrane estrogen receptors (ERs), including membrane subpopulations of ERα and ERβ. In the mid-1990s, several laboratories independently reported the cloning of an orphan G protein-coupled receptor from vascular and cancer cells that was named GPR30. Research published between 2000 and 2005 provided evidence that GPR30 binds and signals via estrogen indicating that this intracellular receptor is involved in rapid, non-genomic estrogen signaling. The receptor has since been designated as the G protein-coupled estrogen receptor (GPER) by the International Union of Pharmacology. The availability of genetic tools such as different lines of GPER knock-out mice, as well as GPER-selective agonists and antagonists has advanced our understanding, but also added some confusion about the new function of this receptor. GPER not only binds estrogens but also other substances, including SERMs, SERDs, and environmental ER activators (endocrine disruptors; xenoestrogens) and also interacts with other proteins. This article represents a summary of a lecture given at the 7(th) International Meeting on Rapid Responses to Steroid Hormones in September 2011 in Axos, Crete, and reviews the current knowledge and questions about GPER-dependent signaling and function. Controversies that have complicated our understanding of GPER, including interactions with human ERα-36 and aldosterone as a potential ligand, will also be discussed.
Collapse
Affiliation(s)
- Matthias Barton
- Molecular Internal Medicine, University of Zurich, LTK Y44 G22, Winterthurerstrasse 190, 8057 Zürich, Switzerland.
| |
Collapse
|