1
|
Wu Y, Xu Y, Xu L. Pharmacological therapy targeting the immune response in atherosclerosis. Int Immunopharmacol 2024; 141:112974. [PMID: 39168023 DOI: 10.1016/j.intimp.2024.112974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024]
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease characterized by the formation of atherosclerotic plaques that consist of numerous cells including smooth muscle cells, endothelial cells, immune cells, and foam cells. The most abundant innate and adaptive immune cells, including neutrophils, monocytes, macrophages, B cells, and T cells, play a pivotal role in the inflammatory response, lipoprotein metabolism, and foam cell formation to accelerate atherosclerotic plaque formation. In this review, we have discussed the underlying mechanisms of activated immune cells in promoting AS and reviewed published clinical trials for the treatment of AS by suppressing immune cell activation. We have also presented some crucial shortcomings of current clinical trials. Lastly, we have discussed the therapeutic potential of novel compounds, including herbal medicine and dietary food, in alleviating AS in animals. Despite these limitations, further clinical trials and experimental studies will enhance our understanding of the mechanisms modulated by immune cells and promote widespread drug use to treat AS by suppressing immune system-induced inflammation.
Collapse
Affiliation(s)
- Yirong Wu
- Department of Cardiology, Hangzhou First People's Hospital, 310006 Zhejiang, China
| | - Yizhou Xu
- Department of Cardiology, Hangzhou First People's Hospital, 310006 Zhejiang, China.
| | - Linhao Xu
- Department of Cardiology, Hangzhou First People's Hospital, 310006 Zhejiang, China; Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Translational Medicine Research Center, Hangzhou First People's Hospital, Hangzhou 310006, Zhejiang, China.
| |
Collapse
|
2
|
Castro R, Adair JH, Mastro AM, Neuberger T, Matters GL. VCAM-1-targeted nanoparticles to diagnose, monitor and treat atherosclerosis. Nanomedicine (Lond) 2024; 19:723-735. [PMID: 38420919 DOI: 10.2217/nnm-2023-0282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024] Open
Abstract
Vascular cell adhesion molecule-1 (VCAM-1) was identified over 2 decades ago as an endothelial adhesion receptor involved in leukocyte recruitment and cell-based immune responses. In atherosclerosis, a chronic inflammatory disease of the blood vessels that is the leading cause of death in the USA, endothelial VCAM-1 is robustly expressed beginning in the early stages of the disease. The interactions of circulating immune cells with VCAM-1 on the activated endothelial cell surface promote the uptake of monocytes and the progression of atherosclerotic lesions in susceptible vessels. Herein, we review the role of VCAM-1 in atherosclerosis and the use of VCAM-1 binding peptides, antibodies and aptamers as targeting agents for nanoplatforms for early detection and treatment of atherosclerotic disease.
Collapse
Affiliation(s)
- Rita Castro
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Pharmaceutical Sciences & Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003, Lisboa, Portugal
| | - James H Adair
- Department of Materials Science, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Pharmacology, The Pennsylvania State University, University Park, PA 16802, USA
| | | | - Thomas Neuberger
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
- Huck Institutes of The Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Gail L Matters
- Department of Biochemistry & Molecular Biology, Penn State College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
3
|
Ferrell M, Wang Z, Anderson JT, Li XS, Witkowski M, DiDonato JA, Hilser JR, Hartiala JA, Haghikia A, Cajka T, Fiehn O, Sangwan N, Demuth I, König M, Steinhagen-Thiessen E, Landmesser U, Tang WHW, Allayee H, Hazen SL. A terminal metabolite of niacin promotes vascular inflammation and contributes to cardiovascular disease risk. Nat Med 2024; 30:424-434. [PMID: 38374343 PMCID: PMC11841810 DOI: 10.1038/s41591-023-02793-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 12/22/2023] [Indexed: 02/21/2024]
Abstract
Despite intensive preventive cardiovascular disease (CVD) efforts, substantial residual CVD risk remains even for individuals receiving all guideline-recommended interventions. Niacin is an essential micronutrient fortified in food staples, but its role in CVD is not well understood. In this study, untargeted metabolomics analysis of fasting plasma from stable cardiac patients in a prospective discovery cohort (n = 1,162 total, n = 422 females) suggested that niacin metabolism was associated with incident major adverse cardiovascular events (MACE). Serum levels of the terminal metabolites of excess niacin, N1-methyl-2-pyridone-5-carboxamide (2PY) and N1-methyl-4-pyridone-3-carboxamide (4PY), were associated with increased 3-year MACE risk in two validation cohorts (US n = 2,331 total, n = 774 females; European n = 832 total, n = 249 females) (adjusted hazard ratio (HR) (95% confidence interval) for 2PY: 1.64 (1.10-2.42) and 2.02 (1.29-3.18), respectively; for 4PY: 1.89 (1.26-2.84) and 1.99 (1.26-3.14), respectively). Phenome-wide association analysis of the genetic variant rs10496731, which was significantly associated with both 2PY and 4PY levels, revealed an association of this variant with levels of soluble vascular adhesion molecule 1 (sVCAM-1). Further meta-analysis confirmed association of rs10496731 with sVCAM-1 (n = 106,000 total, n = 53,075 females, P = 3.6 × 10-18). Moreover, sVCAM-1 levels were significantly correlated with both 2PY and 4PY in a validation cohort (n = 974 total, n = 333 females) (2PY: rho = 0.13, P = 7.7 × 10-5; 4PY: rho = 0.18, P = 1.1 × 10-8). Lastly, treatment with physiological levels of 4PY, but not its structural isomer 2PY, induced expression of VCAM-1 and leukocyte adherence to vascular endothelium in mice. Collectively, these results indicate that the terminal breakdown products of excess niacin, 2PY and 4PY, are both associated with residual CVD risk. They also suggest an inflammation-dependent mechanism underlying the clinical association between 4PY and MACE.
Collapse
Affiliation(s)
- Marc Ferrell
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Systems Biology and Bioinformatics Program, Department of Nutrition, Case Western Reserve University, Cleveland, OH, USA
| | - Zeneng Wang
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - James T Anderson
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Xinmin S Li
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Marco Witkowski
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Cardiology, Angiology and Intensive Care, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany
| | - Joseph A DiDonato
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - James R Hilser
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jaana A Hartiala
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Arash Haghikia
- Department of Cardiology, Angiology and Intensive Care, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Friede Springer Cardiovascular Prevention Center at Charité, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Tomas Cajka
- West Coast Metabolomics Center, University of California, Davis, Davis, CA, USA
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California, Davis, Davis, CA, USA
| | - Naseer Sangwan
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ilja Demuth
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany
| | - Maximilian König
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Ulf Landmesser
- Department of Cardiology, Angiology and Intensive Care, Deutsches Herzzentrum der Charité, Campus Benjamin Franklin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Friede Springer Cardiovascular Prevention Center at Charité, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - W H Wilson Tang
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Hooman Allayee
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Stanley L Hazen
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
4
|
Terasawa M, Zang L, Hiramoto K, Shimada Y, Mitsunaka M, Uchida R, Nishiura K, Matsuda K, Nishimura N, Suzuki K. Oral Administration of Rhamnan Sulfate from Monostroma nitidum Suppresses Atherosclerosis in ApoE-Deficient Mice Fed a High-Fat Diet. Cells 2023; 12:2666. [PMID: 37998401 PMCID: PMC10670814 DOI: 10.3390/cells12222666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/10/2023] [Accepted: 11/16/2023] [Indexed: 11/25/2023] Open
Abstract
Oral administration of rhamnan sulfate (RS), derived from the seaweed Monostroma nitidum, markedly suppresses inflammatory damage in the vascular endothelium and organs of lipopolysaccharide-treated mice. This study aimed to analyze whether orally administered RS inhibits the development of atherosclerosis, a chronic inflammation of the arteries. ApoE-deficient female mice were fed a normal or high-fat diet (HFD) with or without RS for 12 weeks. Immunohistochemical and mRNA analyses of atherosclerosis-related genes were performed. The effect of RS on the migration of RAW264.7 cells was also examined in vitro. RS administration suppressed the increase in blood total cholesterol and triglyceride levels. In the aorta of HFD-fed mice, RS reduced vascular smooth muscle cell proliferation, macrophage accumulation, and elevation of VCAM-1 and inhibited the reduction of Robo4. Increased mRNA levels of Vcam1, Mmp9, and Srebp1 in atherosclerotic areas of HFD-fed mice were also suppressed with RS. Moreover, RS directly inhibited the migration of RAW264.7 cells in vitro. Thus, in HFD-fed ApoE-deficient mice, oral administration of RS ameliorated abnormal lipid metabolism and reduced vascular endothelial inflammation and hyperpermeability, macrophage infiltration and accumulation, and smooth muscle cell proliferation in the arteries leading to atherosclerosis. These results suggest that RS is an effective functional food for the prevention of atherosclerosis.
Collapse
Affiliation(s)
- Masahiro Terasawa
- Konan Chemical Manufacturing Co., Ltd., Kitagomizuka, Kusu-cho, Yokkaichi 510-0103, Japan; (M.T.); (R.U.); (K.N.); (K.M.)
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Minamitamagaki-cho, Suzuka 513-8670, Japan;
| | - Liqing Zang
- Graduate School of Regional Innovation Studies, Mie University, Tsu 514-8507, Japan; (L.Z.); (N.N.)
| | - Keiichi Hiramoto
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Minamitamagaki-cho, Suzuka 513-8670, Japan;
| | - Yasuhito Shimada
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu 514-8507, Japan; (Y.S.); (M.M.)
| | - Mari Mitsunaka
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu 514-8507, Japan; (Y.S.); (M.M.)
| | - Ryota Uchida
- Konan Chemical Manufacturing Co., Ltd., Kitagomizuka, Kusu-cho, Yokkaichi 510-0103, Japan; (M.T.); (R.U.); (K.N.); (K.M.)
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Minamitamagaki-cho, Suzuka 513-8670, Japan;
| | - Kaoru Nishiura
- Konan Chemical Manufacturing Co., Ltd., Kitagomizuka, Kusu-cho, Yokkaichi 510-0103, Japan; (M.T.); (R.U.); (K.N.); (K.M.)
| | - Koichi Matsuda
- Konan Chemical Manufacturing Co., Ltd., Kitagomizuka, Kusu-cho, Yokkaichi 510-0103, Japan; (M.T.); (R.U.); (K.N.); (K.M.)
| | - Norihiro Nishimura
- Graduate School of Regional Innovation Studies, Mie University, Tsu 514-8507, Japan; (L.Z.); (N.N.)
| | - Koji Suzuki
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Minamitamagaki-cho, Suzuka 513-8670, Japan;
| |
Collapse
|
5
|
Billah M, Naz A, Noor R, Bhindi R, Khachigian LM. Early Growth Response-1: Friend or Foe in the Heart? Heart Lung Circ 2023; 32:e23-e35. [PMID: 37024319 DOI: 10.1016/j.hlc.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 04/07/2023]
Abstract
Cardiovascular disease is a major cause of mortality and morbidity worldwide. Early growth response-1 (Egr-1) plays a critical regulatory role in a range of experimental models of cardiovascular diseases. Egr-1 is an immediate-early gene and is upregulated by various stimuli including shear stress, oxygen deprivation, oxidative stress and nutrient deprivation. However, recent research suggests a new, underexplored cardioprotective side of Egr-1. The main purpose of this review is to explore and summarise the dual nature of Egr-1 in cardiovascular pathobiology.
Collapse
Affiliation(s)
- Muntasir Billah
- Department of Cardiology, Kolling Institute of Medical Research, Northern Sydney Local Health District, Sydney, NSW, Australia; Sydney Medical School Northern, The University of Sydney, Sydney, NSW, Australia.
| | - Adiba Naz
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
| | - Rashed Noor
- School of Environmental and Life Sciences, Independent University Bangladesh, Dhaka, Bangladesh
| | - Ravinay Bhindi
- Department of Cardiology, Kolling Institute of Medical Research, Northern Sydney Local Health District, Sydney, NSW, Australia; Sydney Medical School Northern, The University of Sydney, Sydney, NSW, Australia
| | - Levon M Khachigian
- Vascular Biology and Translational Research, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
6
|
Bunet R, Roy-Cardinal MH, Ramani H, Cleret-Buhot A, Durand M, Chartrand-Lefebvre C, Routy JP, Thomas R, Trottier B, Ancuta P, Hanna DB, Landay AL, Cloutier G, Tremblay CL, El-Far M. Differential Impact of IL-32 Isoforms on the Functions of Coronary Artery Endothelial Cells: A Potential Link with Arterial Stiffness and Atherosclerosis. Viruses 2023; 15:700. [PMID: 36992409 PMCID: PMC10052544 DOI: 10.3390/v15030700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/24/2023] [Accepted: 03/04/2023] [Indexed: 03/11/2023] Open
Abstract
Chronic inflammation is associated with higher risk of cardiovascular disease (CVD) in people living with HIV (PLWH). We have previously shown that interleukin-32 (IL-32), a multi-isoform proinflammatory cytokine, is chronically upregulated in PLWH and is linked with CVD. However, the mechanistic roles of the different IL-32 isoforms in CVD are yet to be identified. In this study, we aimed to investigate the potential impact of IL-32 isoforms on coronary artery endothelial cells (CAEC), whose dysfunction represents a major factor for atherosclerosis. Our results demonstrated that the predominantly expressed IL-32 isoforms (IL-32β and IL-32γ) have a selective impact on the production of the proinflammatory cytokine IL-6 by CAEC. Furthermore, these two isoforms induced endothelial cell dysfunction by upregulating the expression of the adhesion molecules ICAM-I and VCAM-I and the chemoattractants CCL-2, CXCL-8 and CXCL-1. IL-32-mediated expression of these chemokines was sufficient to drive monocyte transmigration in vitro. Finally, we demonstrate that IL-32 expression in both PLWH and controls correlates with the carotid artery stiffness, measured by the cumulated lateral translation. These results suggest a role for IL-32-mediated endothelial cell dysfunction in dysregulation of the blood vessel wall and that IL-32 may represent a therapeutic target to prevent CVD in PLWH.
Collapse
Affiliation(s)
- Rémi Bunet
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
| | - Marie-Hélène Roy-Cardinal
- Laboratory of Biorheology and Medical Ultrasonics, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
| | - Hardik Ramani
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
| | - Aurélie Cleret-Buhot
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
| | - Madeleine Durand
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
- Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Carl Chartrand-Lefebvre
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
- Département de Radiologie, Radio-Oncologie et Médecine Nucléaire, Faculté de Médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Jean-Pierre Routy
- Chronic Viral Illness Service, Division of Hematology, McGill University Health Centre, Montréal and Research Institute of McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | - Réjean Thomas
- Clinique Médicale l’Actuel, Montréal, QC H2L 4P9, Canada
| | - Benoît Trottier
- Centre de Médecine Urbaine du Quartier Latin, Montréal, QC H2L 4E9, Canada
| | - Petronela Ancuta
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
| | - David B. Hanna
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Alan L. Landay
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Guy Cloutier
- Laboratory of Biorheology and Medical Ultrasonics, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
- Département de Radiologie, Radio-Oncologie et Médecine Nucléaire, Faculté de Médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada
- Institut de Génie Biomédical, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Cécile L. Tremblay
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, QC H3C 3J7, Canada
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
| | - Mohamed El-Far
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
| |
Collapse
|
7
|
Mauersberger C, Hinterdobler J, Schunkert H, Kessler T, Sager HB. Where the Action Is-Leukocyte Recruitment in Atherosclerosis. Front Cardiovasc Med 2022; 8:813984. [PMID: 35087886 PMCID: PMC8787128 DOI: 10.3389/fcvm.2021.813984] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/15/2021] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is the leading cause of death worldwide and leukocyte recruitment is a key element of this phenomenon, thus allowing immune cells to enter the arterial wall. There, in concert with accumulating lipids, the invading leukocytes trigger a plethora of inflammatory responses which promote the influx of additional leukocytes and lead to the continued growth of atherosclerotic plaques. The recruitment process follows a precise scheme of tethering, rolling, firm arrest, crawling and transmigration and involves multiple cellular and subcellular players. This review aims to provide a comprehensive up-to-date insight into the process of leukocyte recruitment relevant to atherosclerosis, each from the perspective of endothelial cells, monocytes and macrophages, neutrophils, T lymphocytes and platelets. In addition, therapeutic options targeting leukocyte recruitment into atherosclerotic lesions-or potentially arising from the growing body of insights into its precise mechanisms-are highlighted.
Collapse
Affiliation(s)
- Carina Mauersberger
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Julia Hinterdobler
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Heribert Schunkert
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Thorsten Kessler
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Hendrik B. Sager
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
8
|
Meng Q, Lu Q, Zhang Z, Liu J, Lou Y, Wang Y, Liu J. Nesfatin-1 inhibits free fatty acid (FFA)-induced endothelial inflammation via Gfi1/NF-κB signaling. Biosci Biotechnol Biochem 2021; 86:47-55. [PMID: 34724039 DOI: 10.1093/bbb/zbab186] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/02/2021] [Indexed: 11/14/2022]
Abstract
Nesfatin-1 is a neuropeptide produced in the hypothalamus. It is known that Nesfatin-1 is involved in food uptake, fat storage, and other metabolic regulation. We hypothesized that Nesfatin-1 may play a role in cardiovascular tissue. Free fatty acids (FFAs) are known to be the risk factor for cardiovascular diseases. FFA-mediated endothelial dysfunction is the critical mechanism of many cardiovascular disorders. The present study explores the protective effects of Nesfatin-1 on FFA-induced endothelial inflammation and the underlying mechanism. We found that significantly increased lactate dehydrogenase release and production of inflammatory factors were observed in FFA-treated human aortic endothelial cells (HAECs), accompanied by the enhanced attachment of U937 monocytes to HAECs and upregulated cell adhesion molecule vascular cell adhesion molecule-1, which were dramatically reversed by the treatment with Nesfatin-1. In addition, the promoted level of nuclear regulator NF-κB p65 and transcriptional function of NF-κB in FFA-treated HAECs were greatly suppressed by HAECs. Growth Factor Independent 1 Transcriptional Repressor 1 (Gfi1), an important negative regulator of NF-κB activity, was significantly downregulated in HAECs by FFAs and was upregulated by Nesfatin-1. Lastly, the inhibitory effects of Nesfatin-1 against FFA-induced NF-κB activation and adhesion of U937 monocytes to HAECs were abolished by the knockdown of Gfi1. In conclusion, our data reveal that Nesfatin-1 inhibited FFA-induced endothelial inflammation mediated by the Gfi1/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Qingtao Meng
- Department of Cardiovascular Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Qin Lu
- Department of Cardiology, the Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Zhipeng Zhang
- Department of Cardiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Jiyi Liu
- Department of Cardiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Yu Lou
- Department of Cardiology, the Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yuwei Wang
- Department of Cardiovascular Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Jihong Liu
- Department of Cardiovascular Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| |
Collapse
|
9
|
Belcastro E, Rehman AU, Remila L, Park SH, Gong DS, Anton N, Auger C, Lefebvre O, Goetz JG, Collot M, Klymchenko AS, Vandamme TF, Schini-Kerth VB. Fluorescent nanocarriers targeting VCAM-1 for early detection of senescent endothelial cells. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 34:102379. [PMID: 33713860 DOI: 10.1016/j.nano.2021.102379] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 01/29/2021] [Accepted: 02/20/2021] [Indexed: 10/21/2022]
Abstract
Endothelial senescence has been identified as an early event in the development of endothelial dysfunction, a hallmark of cardiovascular disease. This study developed theranostic nanocarriers (NC) decorated with VCAM-1 antibodies (NC-VCAM-1) in order to target cell surface VCAM-1, which is overexpressed in senescent endothelial cells (ECs) for diagnostic and therapeutic purposes. Incubation of Ang II-induced premature senescent ECs or replicative senescent ECs with NC-VCAM-1 loaded with lipophilic fluorescent dyes showed higher fluorescence signals than healthy EC, which was dependent on the NC size and VCAM-1 antibodies concentration, and not observed following masking of VCAM-1. NC loaded with omega 3 polyunsaturated fatty acid (NC-EPA:DHA6:1) were more effective than native EPA:DHA 6:1 to prevent Ang II-induced VCAM-1 and p53 upregulation, and SA-β-galactosidase activity in coronary artery segments. These theranostic NC might be of interest to evaluate the extent and localization of endothelial senescence and to prevent pro-senescent endothelial responses.
Collapse
Affiliation(s)
- Eugenia Belcastro
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, Faculty of Pharmacy
| | - Asad Ur Rehman
- University of Strasbourg, CNRS, CAMB UMR 7199, Strasbourg, France
| | - Lamia Remila
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, Faculty of Pharmacy
| | - Sin-Hee Park
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, Faculty of Pharmacy
| | - Dal Seong Gong
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, Faculty of Pharmacy
| | - Nicolas Anton
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, Faculty of Pharmacy; University of Strasbourg, CNRS, CAMB UMR 7199, Strasbourg, France
| | - Cyril Auger
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, Faculty of Pharmacy
| | | | | | - Mayeul Collot
- CNRS UMR 7213, Laboratory of Biophotonics and Pharmacology, University of Strasbourg, Strasbourg, France
| | - Andrey S Klymchenko
- CNRS UMR 7213, Laboratory of Biophotonics and Pharmacology, University of Strasbourg, Strasbourg, France
| | - Thierry F Vandamme
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, Faculty of Pharmacy; University of Strasbourg, CNRS, CAMB UMR 7199, Strasbourg, France
| | - Valérie B Schini-Kerth
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, Faculty of Pharmacy.
| |
Collapse
|
10
|
Fledderus J, Vanchin B, Rots MG, Krenning G. The Endothelium as a Target for Anti-Atherogenic Therapy: A Focus on the Epigenetic Enzymes EZH2 and SIRT1. J Pers Med 2021; 11:jpm11020103. [PMID: 33562658 PMCID: PMC7915331 DOI: 10.3390/jpm11020103] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 12/11/2022] Open
Abstract
Endothelial cell inflammatory activation and dysfunction are key events in the pathophysiology of atherosclerosis, and are associated with an elevated risk of cardiovascular events. Yet, therapies specifically targeting the endothelium and atherosclerosis are lacking. Here, we review how endothelial behaviour affects atherogenesis and pose that the endothelium may be an efficacious cellular target for antiatherogenic therapies. We discuss the contribution of endothelial inflammatory activation and dysfunction to atherogenesis and postulate that the dysregulation of specific epigenetic enzymes, EZH2 and SIRT1, aggravate endothelial dysfunction in a pleiotropic fashion. Moreover, we propose that commercially available drugs are available to clinically explore this postulation.
Collapse
Affiliation(s)
- Jolien Fledderus
- Medical Biology Section, Laboratory for Cardiovascular Regenerative Medicine, Department Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713 GZ Groningen, The Netherlands; (J.F.); (B.V.)
| | - Byambasuren Vanchin
- Medical Biology Section, Laboratory for Cardiovascular Regenerative Medicine, Department Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713 GZ Groningen, The Netherlands; (J.F.); (B.V.)
- Department Cardiology, School of Medicine, Mongolian National University of Medical Sciences, Jamyan St 3, Ulaanbaatar 14210, Mongolia
| | - Marianne G. Rots
- Epigenetic Editing, Medical Biology Section, Department Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713 GZ Groningen, The Netherlands;
| | - Guido Krenning
- Medical Biology Section, Laboratory for Cardiovascular Regenerative Medicine, Department Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713 GZ Groningen, The Netherlands; (J.F.); (B.V.)
- Correspondence: ; Tel.: +31-50-361-8043; Fax: +31-50-361-9911
| |
Collapse
|
11
|
Bourgeois R, Girard A, Perrot N, Guertin J, Mitchell PL, Couture C, Gotti C, Bourassa S, Poggio P, Mass E, Capoulade R, Scipione CA, Després AA, Couture P, Droit A, Pibarot P, Boffa MB, Thériault S, Koschinsky ML, Mathieu P, Arsenault BJ. A Comparative Analysis of the Lipoprotein(a) and Low-Density Lipoprotein Proteomic Profiles Combining Mass Spectrometry and Mendelian Randomization. CJC Open 2020; 3:450-459. [PMID: 34027348 PMCID: PMC8129481 DOI: 10.1016/j.cjco.2020.11.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 11/28/2020] [Indexed: 12/30/2022] Open
Abstract
Background Lipoprotein(a) (Lp[a]), which consists of a low-density lipoprotein (LDL) bound to apolipoprotein(a), is one of the strongest genetic risk factors for atherosclerotic cardiovascular diseases. Few studies have performed hypothesis-free direct comparisons of the Lp(a) and the LDL proteomes. Our objectives were to compare the Lp(a) and the LDL proteomic profiles and to evaluate the effect of lifelong exposure to elevated Lp(a) or LDL cholesterol levels on the plasma proteomic profile. Methods We performed a label-free analysis of the Lp(a) and LDL proteomic profiles of healthy volunteers in a discovery (n = 6) and a replication (n = 9) phase. We performed inverse variance weighted Mendelian randomization to document the effect of lifelong exposure to elevated Lp(a) or LDL cholesterol levels on the plasma proteomic profile of participants of the INTERVAL study. Results We identified 15 proteins that were more abundant on Lp(a) compared with LDL (serping1, pi16, itih1, itih2, itih3, pon1, podxl, cd44, cp, ptprg, vtn, pcsk9, igfals, vcam1, and ttr). We found no proteins that were more abundant on LDL compared with Lp(a). After correction for multiple testing, lifelong exposure to elevated LDL cholesterol levels was associated with the variation of 18 plasma proteins whereas Lp(a) did not appear to influence the plasma proteome. Conclusions Results of this study highlight marked differences in the proteome of Lp(a) and LDL as well as in the effect of lifelong exposure to elevated LDL cholesterol or Lp(a) on the plasma proteomic profile.
Collapse
Affiliation(s)
- Raphaëlle Bourgeois
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Arnaud Girard
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Nicolas Perrot
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Jakie Guertin
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Patricia L Mitchell
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada
| | - Christian Couture
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada
| | - Clarisse Gotti
- Proteomics platform of the CHU de Québec, Quebec, Canada
| | | | | | - Elvira Mass
- University of Bonn, Developmental Biology of the Immune System, Life and Medical Sciences Institute (LIMES), Bonn, Germany
| | - Romain Capoulade
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Corey A Scipione
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Audrey-Anne Després
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Patrick Couture
- Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada.,Centre de recherche du CHU de Québec, Quebec, Canada
| | - Arnaud Droit
- Proteomics platform of the CHU de Québec, Quebec, Canada.,Centre de recherche du CHU de Québec, Quebec, Canada
| | - Philippe Pibarot
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Michael B Boffa
- Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Sébastien Thériault
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Marlys L Koschinsky
- Robarts Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Patrick Mathieu
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Surgery, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Benoit J Arsenault
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université Laval, Quebec, Canada
| |
Collapse
|
12
|
ABC Transporters, Cholesterol Efflux, and Implications for Cardiovascular Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1276:67-83. [DOI: 10.1007/978-981-15-6082-8_6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
13
|
Mury P, Chirico EN, Mura M, Millon A, Canet-Soulas E, Pialoux V. Oxidative Stress and Inflammation, Key Targets of Atherosclerotic Plaque Progression and Vulnerability: Potential Impact of Physical Activity. Sports Med 2019; 48:2725-2741. [PMID: 30302720 DOI: 10.1007/s40279-018-0996-z] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Atherosclerosis, a complex cardiovascular disease, is a leading cause of mortality and morbidity worldwide. Oxidative stress and inflammation are both involved in the development of atherosclerotic plaque as they increase the biological processes associated with this pathology, such as endothelial dysfunction and macrophage recruitment and adhesion. Atherosclerotic plaque rupture leading to major ischemic events is the result of vulnerable plaque progression, which is a result of the detrimental effect of oxidative stress and inflammation on risk factors for atherosclerotic plaque rupture, such as intraplaque hemorrhage, neovascularization, and fibrous cap thickness. Thus, both are key targets for primary and secondary interventions. It is well recognized that chronic physical activity attenuates oxidative stress in healthy subjects via the improvement of antioxidant enzyme capacities and inflammation via the enhancement of anti-inflammatory molecules. Moreover, it was recently shown that chronic physical activity could decrease oxidative stress and inflammation in atherosclerotic patients. The aim of this review is to summarize the role of oxidative stress and inflammation in atherosclerosis and the results of therapeutic interventions targeting them in both preclinical and clinical studies. The effects of chronic physical activity on these two key processes are then reviewed in vulnerable atherosclerotic plaques in both coronary and carotid arteries.
Collapse
Affiliation(s)
- Pauline Mury
- Team Vascular Biology and Red Blood Cell, Interuniversity Laboratory of Human Movement Biology, University Claude Bernard Lyon 1, University of Lyon, Faculté de Médecine Lyon Est, 8 Avenue Rockefeller, 69008, Lyon, France.,Laboratory of Excellence GR-Ex, Paris, France
| | - Erica N Chirico
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Mathilde Mura
- Team Vascular Biology and Red Blood Cell, Interuniversity Laboratory of Human Movement Biology, University Claude Bernard Lyon 1, University of Lyon, Faculté de Médecine Lyon Est, 8 Avenue Rockefeller, 69008, Lyon, France.,Laboratory of Excellence GR-Ex, Paris, France
| | - Antoine Millon
- University of Lyon, University Claude Bernard Lyon 1, CarMeN Laboratory, INSERM U1060, Bron, France.,Department of Vascular Surgery, Edouard Herriot Hospital, Lyon, France
| | - Emmanuelle Canet-Soulas
- University of Lyon, University Claude Bernard Lyon 1, CarMeN Laboratory, INSERM U1060, Bron, France
| | - Vincent Pialoux
- Team Vascular Biology and Red Blood Cell, Interuniversity Laboratory of Human Movement Biology, University Claude Bernard Lyon 1, University of Lyon, Faculté de Médecine Lyon Est, 8 Avenue Rockefeller, 69008, Lyon, France. .,Laboratory of Excellence GR-Ex, Paris, France. .,Institut Universitaire de France, Paris, France.
| |
Collapse
|
14
|
Li R, Dong Z, Zhuang X, Liu R, Yan F, Chen Y, Gao X, Shi H. Salidroside prevents tumor necrosis factor-α-induced vascular inflammation by blocking mitogen-activated protein kinase and NF-κB signaling activation. Exp Ther Med 2019; 18:4137-4143. [PMID: 31656544 DOI: 10.3892/etm.2019.8064] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 09/10/2019] [Indexed: 02/07/2023] Open
Abstract
Vascular inflammation is a key factor in the pathogenesis of atherosclerosis. Salidroside is an important active ingredient extracted from the root of the Rhodiola rosea plant, which has been reported to have antioxidative, anti-cancer, neuroprotective and cardioprotective effects. However, the effects of salidroside on vascular inflammation have not been clarified. The purpose of the present study was to investigate the protective effects of salidroside against tumor necrosis factor (TNF)-α-induced vascular inflammation in cardiac microvascular endothelial cells (CMECs), a specific cell type derived from coronary micro-vessels. Over a 24-h period, salidroside did not exert any significant cytotoxicity up to a dose of 100 µM. Additionally, salidroside decreased the expression levels of the cell adhesion molecule vascular cell adhesion molecule-1 (VCAM-1) in TNF-α-stimulated CMECs, thus suppressing monocyte-to-CMEC adhesion. Salidroside also decreased the production of inflammatory cytokines such as interleukin (IL)-1β, IL-6 and monocyte chemotactic protein 1 (MCP-1) in TNF-α-induced CMECs, as well as suppressing TNF-α-activated mitogen-activated protein kinase (MAPK) and NF-κB activation. Since MAPKs and NF-κB both serve notable roles in regulating the expression of VCAM-1, IL-1β, IL-6 and MCP-1, the present study provided a preliminary understanding of the mechanism underlying the protective effects of salidroside. Overall, salidroside alleviated vascular inflammation by mediating MAPK and NF-κB activation in TNF-α-induced CMECs. These results indicated that salidroside may have potential applications as a therapeutic agent against vascular inflammation and atherosclerosis.
Collapse
Affiliation(s)
- Ruoshui Li
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai 200036, P.R. China
| | - Zhen Dong
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Xinyu Zhuang
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai 200036, P.R. China
| | - Rongchen Liu
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai 200036, P.R. China
| | - Fangying Yan
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai 200036, P.R. China
| | - Yufei Chen
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai 200036, P.R. China
| | - Xiufang Gao
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai 200036, P.R. China
| | - Haiming Shi
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai 200036, P.R. China
| |
Collapse
|
15
|
Chai Y, Yin Z, Fan Q, Zhang Z, Ye K, Xu Y, Xiao W, Chai X, Zhu T, Nie H. Protective Effects of Angong Niuhuang Pill on Early Atherosclerosis in ApoE -/- Mice by Reducing the Inflammatory Response. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2019; 2019:9747212. [PMID: 31236126 PMCID: PMC6545748 DOI: 10.1155/2019/9747212] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/25/2019] [Accepted: 05/02/2019] [Indexed: 12/28/2022]
Abstract
Atherosclerosis (AS) is the primary cause of cardiocerebrovascular disease, and inflammation is responsible for the initiation of its pathogenesis. Therefore, targeting inflammatory pathways to prevent AS progression is an ideal strategy. Angong Niuhuang pill (ANP) is a well-known traditional Chinese medicine and has been widely used for thousands of years to treat central nervous system and cardiovascular diseases. In this study, we investigated the role of ANP in reducing inflammation during early AS, using a high-fat diet-induced ApoE-/- mouse model of AS. Compared to those with simvastatin, ANP had no significant effect on serum triglyceride, low-density lipoprotein, and high-density lipoprotein levels. However, it effectively inhibited splenic and vascular inflammation. This agent also reduced the Th17/CD4+T ratio and mRNA expression of IL-6 and increased the Treg/CD4+T ratio and mRNA expression of TGF-β1. Thus, ANP restored Th17/Treg homeostasis in the spleen. It also regulated pro- and anti-inflammatory cytokine expression in the aorta in a similar manner. Further, it downregulated the expression of chemokine receptors (CCR2, CXCR3), their ligands (MCP-1, MCP-2, and MCP-3), and cell adhesion molecules (VCAM-1, ICAM-1) in arterial vessels. These results indicate that ANP can ameliorate the development of early AS, mainly by reducing inflammation instead of acting as an antihyperlipidemic drug.
Collapse
Affiliation(s)
- Yushuang Chai
- Guangzhou Baiyunshan Zhongyi Pharmaceutical Co., Ltd, Guangzhou 510530, China
| | - Zhen Yin
- Guangzhou Baiyunshan Zhongyi Pharmaceutical Co., Ltd, Guangzhou 510530, China
| | - Qinghong Fan
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, Guangdong, China
| | - Zhe Zhang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, Guangdong, China
| | - Kaihe Ye
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, Guangdong, China
| | - Yimin Xu
- Guangzhou Baiyunshan Zhongyi Pharmaceutical Co., Ltd, Guangzhou 510530, China
| | - Wei Xiao
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, Guangdong, China
| | - Xiaomeng Chai
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, Guangdong, China
| | - Tao Zhu
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, Guangdong, China
| | - Hong Nie
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, Guangdong, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, China
| |
Collapse
|
16
|
Day A, Jameson Z, Hyde C, Simbi B, Fowkes R, Lawson C. C-Type Natriuretic Peptide (CNP) Inhibition of Interferon-γ-Mediated Gene Expression in Human Endothelial Cells In Vitro. BIOSENSORS-BASEL 2018; 8:bios8030086. [PMID: 30223437 PMCID: PMC6164118 DOI: 10.3390/bios8030086] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 08/24/2018] [Accepted: 09/11/2018] [Indexed: 01/14/2023]
Abstract
Cardiovascular diseases, including atherosclerosis, now account for more deaths in the Western world than from any other cause. Atherosclerosis has a chronic inflammatory component involving Th1 pro-inflammatory cytokines such as IFN-γ, which is known to induce endothelial cell inflammatory responses. On the other hand CNP, which acts via its receptors to elevate intracellular cGMP, is produced by endothelium and endocardium and is upregulated in atherosclerosis. It is believed to be protective, however its role in vascular inflammation is not well understood. The aim of this study was to investigate the effects of CNP on human endothelial cell inflammatory responses following IFN-γ stimulation. Human umbilical vein endothelial cells were treated with either IFN-γ (10 ng/mL) or CNP (100 nm), or both in combination, followed by analysis by flow cytometry for expression of MHC class I and ICAM-1. IFN-γ significantly increased expression of both molecules, which was significantly inhibited by CNP or the cGMP donor 8-Bromoguanosine 3',5'-cyclic monophosphate (1 µm). CNP also reduced IFN-γ mediated kynurenine generation by the IFN-γ regulated enzyme indoleamine-2,3-deoxygenase (IDO). We conclude that CNP downmodulates IFN-γ induced pro-inflammatory gene expression in human endothelial cells via a cGMP-mediated pathway. Thus, CNP may have a protective role in vascular inflammation and novel therapeutic strategies for CVD based on upregulation of endothelial CNP expression could reduce chronic EC inflammation.
Collapse
Affiliation(s)
- Amy Day
- Cardiovascular and Inflammation Biology Group, Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street London, NW1 0TU, UK.
- Endocrine Signalling Group, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK.
| | - Zoe Jameson
- Cardiovascular and Inflammation Biology Group, Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street London, NW1 0TU, UK.
- Endocrine Signalling Group, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK.
| | - Carolyn Hyde
- Bio-Analysis Centre, London Bioscience Innovation Centre, Royal College Street, London NW1 0NH, UK.
| | - Bigboy Simbi
- Endocrine Signalling Group, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK.
| | - Robert Fowkes
- Endocrine Signalling Group, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK.
| | - Charlotte Lawson
- Cardiovascular and Inflammation Biology Group, Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street London, NW1 0TU, UK.
| |
Collapse
|
17
|
Xu T, Lv Z, Chen Q, Guo M, Wang X, Huang F. Vascular endothelial growth factor over-expressed mesenchymal stem cells-conditioned media ameliorate palmitate-induced diabetic endothelial dysfunction through PI-3K/AKT/m-TOR/eNOS and p38/MAPK signaling pathway. Biomed Pharmacother 2018; 106:491-498. [PMID: 29990837 DOI: 10.1016/j.biopha.2018.06.129] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 06/24/2018] [Accepted: 06/25/2018] [Indexed: 12/17/2022] Open
Abstract
In the pathogenesis of diabetes mellitus (DM), islet microvasculares are severely damaged due to glucolipotoxicity and other reasons. Vascular endothelial growth factor (VEGF) is an indispensable and specific angiogenic factor in the pathogenesis and treatment of diabetic islet microvascular disease. Mesenchymal stem cells (MSCs) are regarded as a promising treatment of diabetes because of their immunosuppressive effect and multipotential differentiation potency. In this study, we tested whether MSCs over-expressing VEGF conditioned medium (MSC-VEGF-CM) could ameliorate pancreatic islet endothelial cells (MS-1) dysfunction induced by a common diabetic inducer palmitate (PA). We found that cell survival and migration were restrained by PA and partly repaired by the pro-protected of MSC-VEGF-CM. Meanwhile, PI-3K/AKT/m-TOR/eNOS and p38/MAPK signaling pathways were also up-regulated. Though apoptosis-related proteins, caspase-3 and caspase-9, had no significantly suppressed between MSC-VEGF-CM and MSC-CM alone, the expression levels of vascular surface factors such as CD31, VE-cadherin, occludin and ICAM-1, were remarkably up-regulated by the pro-protected of MSC-VEGF-CM. Our data suggested that MSC-VEGF-CM had therapeutic effect on the PA-induced dysfunction through the re-activation of PI-3K/AKT/m-TOR/eNOS and p38/MAPK signaling pathways.
Collapse
Affiliation(s)
- Tianwei Xu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Zhengbing Lv
- School of Life Science, Zhejiang Sci-Tech University, Hangzhou, China
| | - Qiuhua Chen
- Intensive Care Unit, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Min Guo
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xufang Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Fengjie Huang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
18
|
Abstract
Over the past decade, studies have repeatedly found single-nucleotide polymorphisms located in the collagen ( COL) 4A1 and COL4A2 genes to be associated with cardiovascular disease (CVD), and the 13q34 locus harboring these genes is one of ~160 genome-wide significant risk loci for coronary artery disease. COL4A1 and COL4A2 encode the α1- and α2-chains of collagen type IV, a major component of basement membranes in various tissues including arteries. Despite the growing body of evidence indicating a role for collagen type IV in CVD, remarkably few studies have aimed to directly investigate such a role. The purpose of this review is to summarize the clinical reports linking 13q34 to coronary artery disease, atherosclerosis, and artery stiffening and to assemble the scattered pieces of evidence from experimental studies based on vascular cells and tissue collectively supporting a role for collagen type IV in atherosclerosis and other macrovascular disease conditions.
Collapse
Affiliation(s)
- L B Steffensen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital , Odense , Denmark.,Centre for Individualized Medicine in Arterial Diseases, Odense University Hospital , Odense , Denmark.,Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark , Odense , Denmark
| | - L M Rasmussen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital , Odense , Denmark.,Centre for Individualized Medicine in Arterial Diseases, Odense University Hospital , Odense , Denmark
| |
Collapse
|
19
|
Russo L, Muturi HT, Ghadieh HE, Wisniewski AM, Morgan EE, Quadri SS, Landesberg GP, Siragy HM, Vazquez G, Scalia R, Gupta R, Najjar SM. Liver-specific rescuing of CEACAM1 reverses endothelial and cardiovascular abnormalities in male mice with null deletion of Ceacam1 gene. Mol Metab 2018; 9:98-113. [PMID: 29396368 PMCID: PMC5870095 DOI: 10.1016/j.molmet.2018.01.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 01/07/2018] [Accepted: 01/14/2018] [Indexed: 12/22/2022] Open
Abstract
Objective Mice with global null mutation of Ceacam1 (Cc1−/−), display impairment of insulin clearance that causes hyperinsulinemia followed by insulin resistance, elevated hepatic de novo lipogenesis, and visceral obesity. In addition, they manifest abnormal vascular permeability and elevated blood pressure. Liver-specific rescuing of Ceacam1 reversed all of the metabolic abnormalities in Cc1−/−liver+ mice. The current study examined whether Cc1−/− male mice develop endothelial and cardiac dysfunction and whether this relates to the metabolic abnormalities caused by defective insulin extraction. Methods and results Myography studies showed reduction of agonist-stimulated nitric oxide production in resistance arterioles in Cc1−/−, but not Cc1−/−liver+ mice. Liver-based rescuing of CEACAM1 also attenuated the abnormal endothelial adhesiveness to circulating leukocytes in parallel to reducing plasma endothelin-1 and recovering plasma nitric oxide levels. Echocardiography studies revealed increased septal wall thickness, cardiac hypertrophy and reduced cardiac performance in Cc1−/−, but not Cc1−/−xliver+ mice. Insulin signaling experiments indicated compromised IRS1/Akt/eNOS pathway leading to lower nitric oxide level, and activated Shc/MAPK pathway leading to more endothelin-1 production in the aortae and hearts of Cc1−/−, but not Cc1−/−xliver+ mice. The increase in the ratio of endothelin-1 receptor A/B indicated an imbalance in the vasomotor activity of Cc1−/− mice, which was normalized in Cc1−/−xliver+ mice. Conclusions The data underscore a critical role for impaired CEACAM1-dependent hepatic insulin clearance pathways and resulting hyperinsulinemia and lipid accumulation in aortae and heart in regulating the cardiovascular function. Mice with global deletion of Ceacam1 gene (Cc1−/−) manifest endothelial dysfunction which is reversed by liver-specific rescuing of CEACAM1. Restoring CEACAM1 expression in the liver reversed cardiac hypertrophy and rescued cardiac performance. Hyperinsulinemia emerging from impaired insulin clearance regulates endothelial and cardiovascular functions.
Collapse
Affiliation(s)
- Lucia Russo
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Harrison T Muturi
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Hilda E Ghadieh
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Alexander M Wisniewski
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Eric E Morgan
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Syed S Quadri
- Department of Endocrinology and Metabolism, College of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Gavin P Landesberg
- Department of Physiology and Cardiovascular Research Center, School of Medicine, Temple University, Philadelphia, PA, USA
| | - Helmy M Siragy
- Department of Endocrinology and Metabolism, College of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Guillermo Vazquez
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Rosario Scalia
- Department of Physiology and Cardiovascular Research Center, School of Medicine, Temple University, Philadelphia, PA, USA
| | - Rajesh Gupta
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Sonia M Najjar
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA; Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA.
| |
Collapse
|
20
|
Fulmer ML, Thewke DP. The Endocannabinoid System and Heart Disease: The Role of Cannabinoid Receptor Type 2. Cardiovasc Hematol Disord Drug Targets 2018; 18:34-51. [PMID: 29412125 PMCID: PMC6020134 DOI: 10.2174/1871529x18666180206161457] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 01/01/2018] [Accepted: 01/01/2018] [Indexed: 12/24/2022]
Abstract
Decades of research has provided evidence for the role of the endocannabinoid system in human health and disease. This versatile system, consisting of two receptors (CB1 and CB2), their endogenous ligands (endocannabinoids), and metabolic enzymes has been implicated in a wide variety of disease states, ranging from neurological disorders to cancer. CB2 has gained much interest for its beneficial immunomodulatory role that can be obtained without eliciting psychotropic effects through CB1. Recent studies have shed light on a protective role of CB2 in cardiovascular disease, an ailment which currently takes more lives each year in Western countries than any other disease or injury. By use of CB2 knockout mice and CB2-selective ligands, knowledge of how CB2 signaling affects atherosclerosis and ischemia has been acquired, providing a major stepping stone between basic science and translational clinical research. Here, we summarize the current understanding of the endocannabinoid system in human pathologies and provide a review of the results from preclinical studies examining its function in cardiovascular disease, with a particular emphasis on possible CB2-targeted therapeutic interventions to alleviate atherosclerosis.
Collapse
Affiliation(s)
- Makenzie L. Fulmer
- Department of Biomedical Sciences, Center for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Douglas P. Thewke
- Department of Biomedical Sciences, Center for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| |
Collapse
|
21
|
Yao L, Bhatta A, Xu Z, Chen J, Toque HA, Chen Y, Xu Y, Bagi Z, Lucas R, Huo Y, Caldwell RB, Caldwell RW. Obesity-induced vascular inflammation involves elevated arginase activity. Am J Physiol Regul Integr Comp Physiol 2017; 313:R560-R571. [PMID: 28835451 PMCID: PMC5792147 DOI: 10.1152/ajpregu.00529.2016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 08/08/2017] [Accepted: 08/08/2017] [Indexed: 01/29/2023]
Abstract
Obesity-induced vascular dysfunction involves pathological remodeling of the visceral adipose tissue (VAT) and increased inflammation. Our previous studies showed that arginase 1 (A1) in endothelial cells (ECs) is critically involved in obesity-induced vascular dysfunction. We tested the hypothesis that EC-A1 activity also drives obesity-related VAT remodeling and inflammation. Our studies utilized wild-type and EC-A1 knockout (KO) mice made obese by high-fat/high-sucrose (HFHS) diet. HFHS diet induced increases in body weight, fasting blood glucose, and VAT expansion. This was accompanied by increased arginase activity and A1 expression in vascular ECs and increased expression of tumor necrosis factor-α (TNF-α), monocyte chemoattractant protein-1 (MCP-1), interleukin-10 (IL-10), vascular cell adhesion molecule-1 (VCAM-1), and intercellular adhesion molecule-1 (ICAM-1) mRNA and protein in both VAT and ECs. HFHS also markedly increased circulating inflammatory monocytes and VAT infiltration by inflammatory macrophages, while reducing reparative macrophages. Additionally, adipocyte size and fibrosis increased and capillary density decreased in VAT. These effects of HFHS, except for weight gain and hyperglycemia, were prevented or reduced in mice lacking EC-A1 or treated with the arginase inhibitor 2-(S)-amino-6-boronohexanoic acid (ABH). In mouse aortic ECs, exposure to high glucose (25 mM) and Na palmitate (200 μM) reduced nitric oxide production and increased A1, TNF-α, VCAM-1, ICAM-1, and MCP-1 mRNA, and monocyte adhesion. Knockout of EC-A1 or ABH prevented these effects. HFHS diet-induced VAT inflammation is mediated by EC-A1 expression/activity. Limiting arginase activity is a possible therapeutic means of controlling obesity-induced vascular and VAT inflammation.
Collapse
Affiliation(s)
- Lin Yao
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Anil Bhatta
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Zhimin Xu
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Jijun Chen
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Haroldo A Toque
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Yongjun Chen
- South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Yimin Xu
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Zsolt Bagi
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Rudolf Lucas
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Yuqing Huo
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
- Department of Cell Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Ruth B Caldwell
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
- Department of Cell Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia
- Veterans Administration Medical Center, Augusta, Georgia; and
| | - R William Caldwell
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia;
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| |
Collapse
|
22
|
Akbar N, Digby JE, Cahill TJ, Tavare AN, Corbin AL, Saluja S, Dawkins S, Edgar L, Rawlings N, Ziberna K, McNeill E, Johnson E, Aljabali AA, Dragovic RA, Rohling M, Belgard TG, Udalova IA, Greaves DR, Channon KM, Riley PR, Anthony DC, Choudhury RP. Endothelium-derived extracellular vesicles promote splenic monocyte mobilization in myocardial infarction. JCI Insight 2017; 2:93344. [PMID: 28878126 PMCID: PMC5621885 DOI: 10.1172/jci.insight.93344] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 07/27/2017] [Indexed: 12/31/2022] Open
Abstract
Transcriptionally activated monocytes are recruited to the heart after acute myocardial infarction (AMI). After AMI in mice and humans, the number of extracellular vesicles (EVs) increased acutely. In humans, EV number correlated closely with the extent of myocardial injury. We hypothesized that EVs mediate splenic monocyte mobilization and program transcription following AMI. Some plasma EVs bear endothelial cell (EC) integrins, and both proinflammatory stimulation of ECs and AMI significantly increased VCAM-1-positive EV release. Injected EC-EVs localized to the spleen and interacted with, and mobilized, splenic monocytes in otherwise naive, healthy animals. Analysis of human plasma EV-associated miRNA showed 12 markedly enriched miRNAs after AMI; functional enrichment analyses identified 1,869 putative mRNA targets, which regulate relevant cellular functions (e.g., proliferation and cell movement). Furthermore, gene ontology termed positive chemotaxis as the most enriched pathway for the miRNA-mRNA targets. Among the identified EV miRNAs, EC-associated miRNA-126-3p and -5p were highly regulated after AMI. miRNA-126-3p and -5p regulate cell adhesion- and chemotaxis-associated genes, including the negative regulator of cell motility, plexin-B2. EC-EV exposure significantly downregulated plexin-B2 mRNA in monocytes and upregulated motility integrin ITGB2. These findings identify EVs as a possible novel signaling pathway by linking ischemic myocardium with monocyte mobilization and transcriptional activation following AMI.
Collapse
Affiliation(s)
- Naveed Akbar
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and
| | - Janet E. Digby
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and
| | - Thomas J. Cahill
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and
| | - Abhijeet N. Tavare
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and
| | - Alastair L. Corbin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Sushant Saluja
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and
| | - Sam Dawkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and
| | - Laurienne Edgar
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and
| | - Nadiia Rawlings
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and
| | - Klemen Ziberna
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and
| | - Eileen McNeill
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and
| | | | | | - Alaa A. Aljabali
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and
| | | | - Mala Rohling
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | | | - Irina A. Udalova
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | | | - Keith M. Channon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and
| | - Paul R. Riley
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | | | - Robin P. Choudhury
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, and
- Acute Vascular Imaging Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
23
|
Eloy JO, Petrilli R, Trevizan LNF, Chorilli M. Immunoliposomes: A review on functionalization strategies and targets for drug delivery. Colloids Surf B Biointerfaces 2017; 159:454-467. [PMID: 28837895 DOI: 10.1016/j.colsurfb.2017.07.085] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 07/26/2017] [Accepted: 07/29/2017] [Indexed: 12/31/2022]
Abstract
Nanoparticles, especially liposomes, have gained prominence in the field of drug delivery for the treatment of human diseases, particularly cancer; they provide several advantages, including controlled drug release, protection of the drug against degradation, improved pharmacokinetics, long circulation, and passive targeting to tumors and inflammatory sites due to the enhanced permeability and retention effect. The functionalization of liposomes with monoclonal antibodies or antibody fragments to generate immunoliposomes has emerged as a promising strategy for targeted delivery to and uptake by cells overexpressing the antigens to these antibodies, with a consequent reduction in side effects. In this review, we address functionalization strategies for the non-covalent and covalent attachment of monoclonal antibodies and their fragments to liposomal surfaces. The main reaction occurs between the sulfhydryl groups of thiolated antibodies and maleimide-containing liposomes. Furthermore, we explore the main targeting possibilities with these ligands for the treatment of a variety of pathologies, including HER2- and EGFR-positive cancers, inflammatory and cardiovascular diseases, infectious diseases, and autoimmune and neurodegenerative diseases, which have not previously been reviewed together. Overall, many studies have shown selective delivery of immunoliposomes to target cells, with promising in vivo results, particularly for cancer treatment. Although clinical trials have been conducted, immunoliposomes have not yet received clinical approval. However, immunoliposomes are promising formulations that are expected to become available for therapeutic use after clinical trials prove their safety and efficacy, and after scaling issues are resolved.
Collapse
Affiliation(s)
- Josimar O Eloy
- School of Pharmaceutical Sciences of Araraquara, São Paulo State University, UNESP, Department of Drugs and Medicines, Araraquara, SP, Brazil.
| | - Raquel Petrilli
- School of Pharmaceutical Sciences of Ribeirão Preto, São Paulo State University, USP, Department of Pharmaceutical Sciences, Ribeirão Preto, SP, Brazil
| | - Lucas Noboru Fatori Trevizan
- School of Pharmaceutical Sciences of Araraquara, São Paulo State University, UNESP, Department of Drugs and Medicines, Araraquara, SP, Brazil
| | - Marlus Chorilli
- School of Pharmaceutical Sciences of Araraquara, São Paulo State University, UNESP, Department of Drugs and Medicines, Araraquara, SP, Brazil
| |
Collapse
|
24
|
Abstract
Molecular imaging offers great potential for noninvasive visualization and quantitation of the cellular and molecular components involved in atherosclerotic plaque stability. In this chapter, we review emerging molecular imaging modalities and approaches for quantitative, noninvasive detection of early biological processes in atherogenesis, including vascular endothelial permeability, endothelial adhesion molecule up-regulation, and macrophage accumulation, with special emphasis on mouse models. We also highlight a number of targeted imaging nanomaterials for assessment of advanced atherosclerotic plaques, including extracellular matrix degradation, proteolytic enzyme activity, and activated platelets using mouse models of atherosclerosis. The potential for clinical translation of molecular imaging nanomaterials for assessment of atherosclerotic plaque biology, together with multimodal approaches is also discussed.
Collapse
|
25
|
Rohart F, Mason EA, Matigian N, Mosbergen R, Korn O, Chen T, Butcher S, Patel J, Atkinson K, Khosrotehrani K, Fisk NM, Lê Cao KA, Wells CA. A molecular classification of human mesenchymal stromal cells. PeerJ 2016; 4:e1845. [PMID: 27042394 PMCID: PMC4811172 DOI: 10.7717/peerj.1845] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/03/2016] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stromal cells (MSC) are widely used for the study of mesenchymal tissue repair, and increasingly adopted for cell therapy, despite the lack of consensus on the identity of these cells. In part this is due to the lack of specificity of MSC markers. Distinguishing MSC from other stromal cells such as fibroblasts is particularly difficult using standard analysis of surface proteins, and there is an urgent need for improved classification approaches. Transcriptome profiling is commonly used to describe and compare different cell types; however, efforts to identify specific markers of rare cellular subsets may be confounded by the small sample sizes of most studies. Consequently, it is difficult to derive reproducible, and therefore useful markers. We addressed the question of MSC classification with a large integrative analysis of many public MSC datasets. We derived a sparse classifier (The Rohart MSC test) that accurately distinguished MSC from non-MSC samples with >97% accuracy on an internal training set of 635 samples from 41 studies derived on 10 different microarray platforms. The classifier was validated on an external test set of 1,291 samples from 65 studies derived on 15 different platforms, with >95% accuracy. The genes that contribute to the MSC classifier formed a protein-interaction network that included known MSC markers. Further evidence of the relevance of this new MSC panel came from the high number of Mendelian disorders associated with mutations in more than 65% of the network. These result in mesenchymal defects, particularly impacting on skeletal growth and function. The Rohart MSC test is a simple in silico test that accurately discriminates MSC from fibroblasts, other adult stem/progenitor cell types or differentiated stromal cells. It has been implemented in the www.stemformatics.org resource, to assist researchers wishing to benchmark their own MSC datasets or data from the public domain. The code is available from the CRAN repository and all data used to generate the MSC test is available to download via the Gene Expression Omnibus or the Stemformatics resource.
Collapse
Affiliation(s)
- Florian Rohart
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- The University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Elizabeth A. Mason
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
| | - Nicholas Matigian
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- The University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Rowland Mosbergen
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- Department of Anatomy and Neuroscience, Faculty of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Othmar Korn
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
| | - Tyrone Chen
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- Department of Anatomy and Neuroscience, Faculty of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Suzanne Butcher
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- Department of Anatomy and Neuroscience, Faculty of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Jatin Patel
- The University of Queensland Centre for Clinical Research, University of Queensland, Brisbane, Queensland, Australia
| | - Kerry Atkinson
- The University of Queensland Centre for Clinical Research, University of Queensland, Brisbane, Queensland, Australia
| | - Kiarash Khosrotehrani
- The University of Queensland Centre for Clinical Research, University of Queensland, Brisbane, Queensland, Australia
- Centre for Advanced Prenatal Care, Royal Brisbane & Women’s Hospital, Brisbane, Queensland, Australia
| | - Nicholas M. Fisk
- The University of Queensland Centre for Clinical Research, University of Queensland, Brisbane, Queensland, Australia
- Centre for Advanced Prenatal Care, Royal Brisbane & Women’s Hospital, Brisbane, Queensland, Australia
| | - Kim-Anh Lê Cao
- The University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Christine A. Wells
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- Department of Anatomy and Neuroscience, Faculty of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
26
|
Denys A, Clavel G, Lemeiter D, Schischmanoff O, Boissier MC, Semerano L. Aortic VCAM-1: an early marker of vascular inflammation in collagen-induced arthritis. J Cell Mol Med 2016; 20:855-63. [PMID: 26859834 PMCID: PMC4831368 DOI: 10.1111/jcmm.12790] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 12/13/2015] [Indexed: 12/16/2022] Open
Abstract
Cardiovascular disease (CVD) is a major cause of morbidity and mortality in rheumatoid arthritis (RA). There are limited experimental data on vascular involvement in arthritis models. To study the link between CVD and inflammation in RA, we developed a model of vascular dysfunction and articular inflammation by collagen‐induced arthritis (CIA) in C57Bl/6 (B6) mice. We studied the expression of vascular inflammatory markers in CIA with and without concomitant hyperlipidic diet (HD). Collagen‐induced arthritis was induced with intradermal injection of chicken type‐II collagen followed by a boost 21 days later. Mice with and without CIA were fed a standard diet or an HD for 12 weeks starting from the day of the boost. Arthritis severity was evaluated with a validated clinical score. Aortic mRNA levels of vascular cell adhesion molecule‐1 (VCAM‐1), inducible nitric oxide synthase (iNOS) and interleukin‐17 were analysed by quantitative RT‐PCR. Vascular cell adhesion molecule‐1 localization in the aortic sinus was determined by immunohistochemistry. Atherosclerotic plaque presence was assessed in aortas. Collagen‐induced arthritis was associated with increased expression of VCAM‐1, independent of diet. VCAM‐1 overexpression was detectable as early as 4 weeks after collagen immunization and persisted after 15 weeks. The HD induced atheroma plaque formation and aortic iNOS expression regardless of CIA. Concomitant CIA and HD had no additive effect on atheroma or VCAM‐1 or iNOS expression. CIA and an HD diet induced a distinct and independent expression of large‐vessel inflammation markers in B6 mice. This model may be relevant for the study of CVD in RA.
Collapse
Affiliation(s)
- Anne Denys
- Inserm UMR 1125, Bobigny, France.,Sorbonne Paris Cité - Université Paris 13, Bobigny, France
| | - Gaëlle Clavel
- Inserm UMR 1125, Bobigny, France.,Department of Internal Medicine, Fondation Rothschild, Paris, France
| | - Delphine Lemeiter
- Inserm UMR 1125, Bobigny, France.,Sorbonne Paris Cité - Université Paris 13, Bobigny, France
| | - Olivier Schischmanoff
- Sorbonne Paris Cité - Université Paris 13, Bobigny, France.,Inserm UMR 978, Bobigny, France
| | - Marie-Christophe Boissier
- Inserm UMR 1125, Bobigny, France.,Sorbonne Paris Cité - Université Paris 13, Bobigny, France.,Assistance Publique - Hôpitaux de Paris (AP-HP) Groupe hospitalier Avicenne - Jean Verdier - René Muret, Service de Rhumatologie, Bobigny, France
| | - Luca Semerano
- Inserm UMR 1125, Bobigny, France.,Sorbonne Paris Cité - Université Paris 13, Bobigny, France.,Assistance Publique - Hôpitaux de Paris (AP-HP) Groupe hospitalier Avicenne - Jean Verdier - René Muret, Service de Rhumatologie, Bobigny, France
| |
Collapse
|
27
|
Inhibitory Effect of a French Maritime Pine Bark Extract-Based Nutritional Supplement on TNF-α-Induced Inflammation and Oxidative Stress in Human Coronary Artery Endothelial Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:260530. [PMID: 26664450 PMCID: PMC4664804 DOI: 10.1155/2015/260530] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 11/17/2022]
Abstract
Oxidative stress and inflammation, leading to endothelial dysfunction, contribute to the pathogenesis of atherosclerosis. The popularity of natural product supplements has increased in recent years, especially those with purported anti-inflammatory and/or antioxidant effects. The efficacy and mechanism of many of these products are not yet well understood. In this study, we tested the antioxidant and anti-inflammatory effects of a supplement, HIPER Health Supplement (HIPER), on cytokine-induced inflammation and oxidative stress in human coronary artery endothelial cells (HCAECs). HIPER is a mixture of French maritime pine bark extract (PBE), honey, aloe vera, and papaya extract. Treatment for 24 hours with HIPER reduced TNF-α-induced reactive oxygen species (ROS) generation that was associated with decreased NADPH oxidase 4 and increased superoxide dismutase-1 expression. HIPER inhibited TNF-α induced monocyte adhesion to HCAECs that was in keeping with decreased expression of vascular cell adhesion molecule-1 and intercellular cell adhesion molecule-1 and decreased nuclear factor-kappa B (NF-κB) activation. Further investigation of mechanism showed HIPER reduced TNF-α induced IκBα and p38 and MEK1/2 MAP kinases phosphorylation. Our findings show that HIPER has potent inhibitory effects on HCAECs inflammatory and oxidative stress responses that may protect against endothelial dysfunction that underlies early atherosclerotic lesion formation.
Collapse
|
28
|
Zhou X, Song G, Zhang X, Liang X, Li Q, Zhang J, Zhou Y. Beneficial effects of crude extract ofEupatorium lindleyanumDC. in hyperlipidemia and atherosclerosis. BIOTECHNOL BIOTEC EQ 2015. [DOI: 10.1080/13102818.2015.1088796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
29
|
Zuo P, Zhou Q, Zuo Z, Wang X, Chen L, Ma G. Effects of the factor Xa inhibitor, fondaparinux, on the stability of atherosclerotic lesions in apolipoprotein E-deficient mice. Circ J 2015; 79:2499-508. [PMID: 26346031 DOI: 10.1253/circj.cj-15-0285] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Atherosclerosis is a progressive inflammatory disease that can lead to sudden cardiac events by plaque rupture and subsequent thrombosis. Factor Xa (FXa) not only occupies a crucial position in the coagulation cascade responsible for thrombin generation, but also has pro-inflammatory effects. The hypothesis that Fondaparinux, the selective FXa inhibitor, attenuates plaque progression and promotes stability of atherosclerotic lesions was assessed. METHODS AND RESULTS Fondaparinux (5 mg/kg body weight/day) or 0.9% saline was intraperitoneally administered for 4 weeks to apolipoprotein E-deficient mice (n=12 per group) with established atherosclerotic lesions in the innominate arteries. Fondaparinux did not remarkably decrease the progression of atherosclerosis development in apolipoprotein E-deficient mice, but increased the thickness of fibrous cap (P=0.049) and decreased the ratio of necrotic core (P=0.001) significantly. Moreover, Fondaparinux reduced the staining against Mac-2 (P=0.017), α-SMA (P=0.002), protease-activated receptor (PAR)-1 (P=0.001), PAR-2 (P=0.003), CD-31 (P=0.024), MMP-9 (P=0.000), MMP-13(P=0.011), VCAM-1 (P=0.041) and the mRNA expression of inflammatory mediators (P<0.05) significantly, such as interleukin (IL)-6, MCP-1, IFN-γ, TNF-α, IL-10 and Egr-1. CONCLUSIONS Fondaparinux, the selective FXa inhibitor, can promote the stability of atherosclerotic lesions in apolipoprotein E-deficient mice, possibly through inhibiting expression of the inflammatory mediators in plaque and reduced synthesis of MMP-9 and MMP-13.
Collapse
Affiliation(s)
- Pengfei Zuo
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University
| | | | | | | | | | | |
Collapse
|
30
|
Marschall ALJ, Single FN, Schlarmann K, Bosio A, Strebe N, van den Heuvel J, Frenzel A, Dübel S. Functional knock down of VCAM1 in mice mediated by endoplasmatic reticulum retained intrabodies. MAbs 2015; 6:1394-401. [PMID: 25484057 PMCID: PMC4622715 DOI: 10.4161/mabs.34377] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Functional knockdowns mediated by endoplasmatic reticulum-retained antibodies (ER intrabodies) are a promising tool for research because they allow functional interference on the protein level. We demonstrate for the first time that ER intrabodies can induce a knock-down phenotype in mice. Surface VCAM1 was suppressed in bone marrow of heterozygous and homozygous ER intrabody mice (iER-VCAM1 mice). iER-VCAM1 mice did not have a lethal phenotype, in contrast to the constitutive knockout of VCAM1, but adult mice exhibited physiological effects in the form of aberrant distribution of immature B-cells in blood and bone marrow. The capability to regulate knock-down strength may spark a new approach for the functional study of membrane and plasma proteins, which may especially be valuable for generating mouse models that more closely resemble disease states than classic knockouts do.
Collapse
Affiliation(s)
- Andrea L J Marschall
- a Technische Universität Braunschweig; Institute of Biochemistry; Biotechnology and Bioinformatics ; Braunschweig , Germany
| | | | | | | | | | | | | | | |
Collapse
|
31
|
LINDHOLM EE, AUNE E, SELJEFLOT I, OTTERSTAD JE, KIRKEBØEN KA. Biomarkers of inflammation in major vascular surgery: a prospective randomised trial. Acta Anaesthesiol Scand 2015; 59:773-87. [PMID: 25626738 DOI: 10.1111/aas.12466] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 12/10/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND Surgery induces inflammation and pro-inflammatory cytokines are associated with post-operative complications. In cardiac surgery, it has been shown that volatile anaesthetics have cardioprotective properties. We explored whether sevoflurane affects the pro-inflammatory response favourably compared with total intravenous anaesthesia (TIVA) after surgery. METHODS We measured monocyte chemotactic protein 1 (MCP-1), matrix metalloproteinase 9 (MMP-9), C-reactive protein (CRP), vascular cell adhesion molecule 1 (VCAM-1), interleukin (IL)-6 and IL-8 perioperatively and evaluated if the anaesthetic regimen affected these mediators. Our hypothesis was that sevoflurane-based anaesthesia is associated with a reduced release of biomarkers of inflammation compared with TIVA with propofol/remifentanil. RESULTS In the total population, MCP-1, MMP-9, IL-6 and IL-8 increased 30 min after arrival intensive care unit, compared with before surgery (P < 0.001), whereas CRP and VCAM-1 transiently declined (P < 0.001). From 30 min after arrival intensive care unit to 1st post-operative day, MCP-1 and IL-6 levels declined (P < 0.001), CRP and VCAM-1 increased (P < 0.001), whereas MMP-9 and IL-8 were not significantly altered. Pre-operatively there were no significant differences in any variables between the two anaesthetic groups. Lower levels of MCP-1 and IL-8 (P < 0.001) and higher levels of IL-6 and MMP-9 (P = 0.003) were found in the sevoflurane group, compared with the TIVA group 30 min post-operatively. CRP and VCAM-1 levels did not differ. There were no significant differences between the two anaesthetic groups before surgery or at 1st post-operative day. CONCLUSION We found an inflammatory response during the observation period, which was modified by the anaesthetic regimen in the early phase. This short-lasting difference is probably too short to support a cardioprotective effect of sevoflurane compared with TIVA in open abdominal aortic surgery.
Collapse
Affiliation(s)
- E. E. LINDHOLM
- Department of Anaesthesiology; Vestfold Hospital Trust; Tønsberg Norway
| | - E. AUNE
- Department of Cardiology; Vestfold Hospital Trust; Tønsberg Norway
| | - I. SELJEFLOT
- Center for Clinical Heart Research; Department of Cardiology; Oslo University Hospital; Oslo Norway
- Faculty of Medicine; University of Oslo; Oslo Norway
| | - J. E. OTTERSTAD
- Department of Cardiology; Vestfold Hospital Trust; Tønsberg Norway
| | - K. A. KIRKEBØEN
- Department of Anaesthesiology; Oslo University Hospital; Oslo Norway
- Faculty of Medicine; University of Oslo; Oslo Norway
| |
Collapse
|
32
|
Kim H, Kee PH, Rim Y, Moody MR, Klegerman ME, Vela D, Huang SL, McPherson DD, Laing ST. Nitric Oxide-Enhanced Molecular Imaging of Atheroma using Vascular Cellular Adhesion Molecule 1-Targeted Echogenic Immunoliposomes. ULTRASOUND IN MEDICINE & BIOLOGY 2015; 41:1701-1710. [PMID: 25819469 PMCID: PMC4426087 DOI: 10.1016/j.ultrasmedbio.2015.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 12/19/2014] [Accepted: 02/09/2015] [Indexed: 06/04/2023]
Abstract
The aim of this study was to determine whether pre-treatment with nitric oxide-loaded echogenic liposomes (NO-ELIP) plus ultrasound can improve highlighting by molecularly targeted (anti-vascular cell adhesion molecule 1 [VCAM-1]) ELIP of atheroma components. Atherosclerotic animals were treated with anti-VCAM-1-ELIP or immunoglobulin (IgG)-ELIP. Each group was selected at random to receive pre-treatment with standard ELIP plus ultrasound, NO-ELIP without ultrasound and NO-ELIP plus ultrasound. Intravascular ultrasound highlighting data for the same arterial segments were collected before and after treatment. Pre-treatment with NO-ELIP plus ultrasound resulted in a significant increase in acoustic enhancement by anti-VCAM-1-ELIP (21.3 ± 1.5% for gray-scale value, 53.9 ± 3.1% for radiofrequency data; p < 0.001 vs. IgG-ELIP, p < 0.05 vs. pre-treatment with standard ELIP plus ultrasound or NO-ELIP without ultrasound). NO-ELIP plus ultrasound can improve highlighting of atheroma by anti-VCAM-1 ELIP. This NO pre-treatment strategy may be useful in optimizing contrast agent delivery to the vascular wall for both diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Hyunggun Kim
- Division of Cardiology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Patrick H Kee
- Division of Cardiology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Yonghoon Rim
- Division of Cardiology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Melanie R Moody
- Division of Cardiology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Melvin E Klegerman
- Division of Cardiology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Deborah Vela
- Department of Pathology, Texas Heart Institute, Houston, Texas, USA
| | - Shao-Ling Huang
- Division of Cardiology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - David D McPherson
- Division of Cardiology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Susan T Laing
- Division of Cardiology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA.
| |
Collapse
|
33
|
Milstone DS, Ilyama M, Chen M, O'Donnell P, Davis VM, Plutzky J, Brown JD, Haldar SM, Siu A, Lau AC, Zhu SN, Basheer MF, Collins T, Jongstra-Bilen J, Cybulsky MI. Differential role of an NF-κB transcriptional response element in endothelial versus intimal cell VCAM-1 expression. Circ Res 2015; 117:166-77. [PMID: 26034041 DOI: 10.1161/circresaha.117.306666] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 05/21/2015] [Indexed: 12/22/2022]
Abstract
RATIONALE Human and murine Vcam1 promoters contain 2 adjacent nuclear factor-κB (NF-κB)-binding elements. Both are essential for cytokine-induced transcription of transiently transfected promoter-reporter constructs. However, the relevance of these insights to regulation of the endogenous Vcam1 gene and to pathophysiological processes in vivo remained unknown. OBJECTIVE Determine the role of the 5' NF-κB-binding element in expression of the endogenous Vcam1 gene. METHODS AND RESULTS Homologous recombination in embryonic stem cells was used to inactivate the 5' NF-κB element in the Vcam1 promoter and alter 3 nucleotides in the 5' untranslated region to allow direct comparison of wild-type versus mutant allele RNA expression and chromatin configuration in heterozygous mice. Systemic treatment with inflammatory cytokines or endotoxin (lipopolysaccharide) induced lower expression of the mutant allele relative to wild-type by endothelial cells in the aorta, heart, and lungs. The mutant allele also showed lower endothelial expression in 2-week atherosclerotic lesions in Vcam1 heterozygous/low-density lipoprotein receptor-deficient mice fed a cholesterol-rich diet. In vivo chromatin immunoprecipitation assays of heart showed diminished lipopolysaccharide-induced association of RNA polymerase 2 and NF-κB p65 with the mutant promoter. In contrast, expression of mutant and wild-type alleles was comparable in intimal cells of wire-injured carotid artery and 4- to 12-week atherosclerotic lesions. CONCLUSIONS This study highlights differences between in vivo and in vitro promoter analyses, and reveals a differential role for a NF-κB transcriptional response element in endothelial vascular cell adhesion molecule-1 expression induced by inflammatory cytokines or a cholesterol-rich diet versus intimal cell expression in atherosclerotic lesions and injured arteries.
Collapse
Affiliation(s)
- David S Milstone
- From the Vascular Research Division, Department of Pathology, Center for Excellence in Vascular Biology (D.S.M., P.O.D., V.M.D., T.C.) and Cardiovascular Division (J.P., J.D.B.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Advanced Diagnostics Division, Toronto General Research Institute, University Health Network Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Geriatric Medicine, Kyoto University Hospital, Kyoto, Japan (M.I.); Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (S.M.H.); and Department of Pathology, Children's Hospital and Harvard Medical School, Boston, MA (T.C.).
| | - Motoi Ilyama
- From the Vascular Research Division, Department of Pathology, Center for Excellence in Vascular Biology (D.S.M., P.O.D., V.M.D., T.C.) and Cardiovascular Division (J.P., J.D.B.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Advanced Diagnostics Division, Toronto General Research Institute, University Health Network Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Geriatric Medicine, Kyoto University Hospital, Kyoto, Japan (M.I.); Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (S.M.H.); and Department of Pathology, Children's Hospital and Harvard Medical School, Boston, MA (T.C.)
| | - Mian Chen
- From the Vascular Research Division, Department of Pathology, Center for Excellence in Vascular Biology (D.S.M., P.O.D., V.M.D., T.C.) and Cardiovascular Division (J.P., J.D.B.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Advanced Diagnostics Division, Toronto General Research Institute, University Health Network Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Geriatric Medicine, Kyoto University Hospital, Kyoto, Japan (M.I.); Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (S.M.H.); and Department of Pathology, Children's Hospital and Harvard Medical School, Boston, MA (T.C.)
| | - Peter O'Donnell
- From the Vascular Research Division, Department of Pathology, Center for Excellence in Vascular Biology (D.S.M., P.O.D., V.M.D., T.C.) and Cardiovascular Division (J.P., J.D.B.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Advanced Diagnostics Division, Toronto General Research Institute, University Health Network Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Geriatric Medicine, Kyoto University Hospital, Kyoto, Japan (M.I.); Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (S.M.H.); and Department of Pathology, Children's Hospital and Harvard Medical School, Boston, MA (T.C.)
| | - Vannessa M Davis
- From the Vascular Research Division, Department of Pathology, Center for Excellence in Vascular Biology (D.S.M., P.O.D., V.M.D., T.C.) and Cardiovascular Division (J.P., J.D.B.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Advanced Diagnostics Division, Toronto General Research Institute, University Health Network Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Geriatric Medicine, Kyoto University Hospital, Kyoto, Japan (M.I.); Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (S.M.H.); and Department of Pathology, Children's Hospital and Harvard Medical School, Boston, MA (T.C.)
| | - Jorge Plutzky
- From the Vascular Research Division, Department of Pathology, Center for Excellence in Vascular Biology (D.S.M., P.O.D., V.M.D., T.C.) and Cardiovascular Division (J.P., J.D.B.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Advanced Diagnostics Division, Toronto General Research Institute, University Health Network Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Geriatric Medicine, Kyoto University Hospital, Kyoto, Japan (M.I.); Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (S.M.H.); and Department of Pathology, Children's Hospital and Harvard Medical School, Boston, MA (T.C.)
| | - Jonathan D Brown
- From the Vascular Research Division, Department of Pathology, Center for Excellence in Vascular Biology (D.S.M., P.O.D., V.M.D., T.C.) and Cardiovascular Division (J.P., J.D.B.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Advanced Diagnostics Division, Toronto General Research Institute, University Health Network Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Geriatric Medicine, Kyoto University Hospital, Kyoto, Japan (M.I.); Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (S.M.H.); and Department of Pathology, Children's Hospital and Harvard Medical School, Boston, MA (T.C.)
| | - Saptarsi M Haldar
- From the Vascular Research Division, Department of Pathology, Center for Excellence in Vascular Biology (D.S.M., P.O.D., V.M.D., T.C.) and Cardiovascular Division (J.P., J.D.B.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Advanced Diagnostics Division, Toronto General Research Institute, University Health Network Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Geriatric Medicine, Kyoto University Hospital, Kyoto, Japan (M.I.); Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (S.M.H.); and Department of Pathology, Children's Hospital and Harvard Medical School, Boston, MA (T.C.)
| | - Allan Siu
- From the Vascular Research Division, Department of Pathology, Center for Excellence in Vascular Biology (D.S.M., P.O.D., V.M.D., T.C.) and Cardiovascular Division (J.P., J.D.B.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Advanced Diagnostics Division, Toronto General Research Institute, University Health Network Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Geriatric Medicine, Kyoto University Hospital, Kyoto, Japan (M.I.); Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (S.M.H.); and Department of Pathology, Children's Hospital and Harvard Medical School, Boston, MA (T.C.)
| | - Andrew C Lau
- From the Vascular Research Division, Department of Pathology, Center for Excellence in Vascular Biology (D.S.M., P.O.D., V.M.D., T.C.) and Cardiovascular Division (J.P., J.D.B.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Advanced Diagnostics Division, Toronto General Research Institute, University Health Network Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Geriatric Medicine, Kyoto University Hospital, Kyoto, Japan (M.I.); Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (S.M.H.); and Department of Pathology, Children's Hospital and Harvard Medical School, Boston, MA (T.C.)
| | - Su-Ning Zhu
- From the Vascular Research Division, Department of Pathology, Center for Excellence in Vascular Biology (D.S.M., P.O.D., V.M.D., T.C.) and Cardiovascular Division (J.P., J.D.B.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Advanced Diagnostics Division, Toronto General Research Institute, University Health Network Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Geriatric Medicine, Kyoto University Hospital, Kyoto, Japan (M.I.); Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (S.M.H.); and Department of Pathology, Children's Hospital and Harvard Medical School, Boston, MA (T.C.)
| | - Mayada F Basheer
- From the Vascular Research Division, Department of Pathology, Center for Excellence in Vascular Biology (D.S.M., P.O.D., V.M.D., T.C.) and Cardiovascular Division (J.P., J.D.B.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Advanced Diagnostics Division, Toronto General Research Institute, University Health Network Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Geriatric Medicine, Kyoto University Hospital, Kyoto, Japan (M.I.); Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (S.M.H.); and Department of Pathology, Children's Hospital and Harvard Medical School, Boston, MA (T.C.)
| | - Tucker Collins
- From the Vascular Research Division, Department of Pathology, Center for Excellence in Vascular Biology (D.S.M., P.O.D., V.M.D., T.C.) and Cardiovascular Division (J.P., J.D.B.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Advanced Diagnostics Division, Toronto General Research Institute, University Health Network Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Geriatric Medicine, Kyoto University Hospital, Kyoto, Japan (M.I.); Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (S.M.H.); and Department of Pathology, Children's Hospital and Harvard Medical School, Boston, MA (T.C.)
| | - Jenny Jongstra-Bilen
- From the Vascular Research Division, Department of Pathology, Center for Excellence in Vascular Biology (D.S.M., P.O.D., V.M.D., T.C.) and Cardiovascular Division (J.P., J.D.B.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Advanced Diagnostics Division, Toronto General Research Institute, University Health Network Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Geriatric Medicine, Kyoto University Hospital, Kyoto, Japan (M.I.); Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (S.M.H.); and Department of Pathology, Children's Hospital and Harvard Medical School, Boston, MA (T.C.)
| | - Myron I Cybulsky
- From the Vascular Research Division, Department of Pathology, Center for Excellence in Vascular Biology (D.S.M., P.O.D., V.M.D., T.C.) and Cardiovascular Division (J.P., J.D.B.), Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Advanced Diagnostics Division, Toronto General Research Institute, University Health Network Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada (M.I., M.C., A.S., A.C.L., S.-N.Z., M.F.B., J.J.-B., M.I.C.); Department of Geriatric Medicine, Kyoto University Hospital, Kyoto, Japan (M.I.); Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH (S.M.H.); and Department of Pathology, Children's Hospital and Harvard Medical School, Boston, MA (T.C.).
| |
Collapse
|
34
|
Serum Levels of IL-1 β , IL-6, TGF- β , and MMP-9 in Patients Undergoing Carotid Artery Stenting and Regulation of MMP-9 in a New In Vitro Model of THP-1 Cells Activated by Stenting. Mediators Inflamm 2015; 2015:956082. [PMID: 26113783 PMCID: PMC4465715 DOI: 10.1155/2015/956082] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 05/07/2015] [Accepted: 05/12/2015] [Indexed: 11/19/2022] Open
Abstract
Inflammation plays an important role in the pathophysiological process after carotid artery stenting (CAS). Monocyte is a significant source of inflammatory cytokines in vascular remodeling. Telmisartan could reduce inflammation. In our study, we first found that, after CAS, the serum IL-1β, IL-6, TGF-β, and MMP-9 levels were significantly increased, but only MMP-9 level was elevated no less than 3 months. Second, we established a new in vitro model, where THP-1 monocytes were treated with the supernatants of human umbilical vein endothelial cells (HUVECs) that were scratched by pipette tips, which mimics monocytes activated by mechanical injury of stenting. The treatment enhanced THP-1 cell adhesion, migration and invasion ability, and the phosphorylation of ERK1/2 and Elk-1 and MMP-9 expression were significantly increased. THP-1 cells pretreated with PD98095 (ERK1/2 inhibitor) attenuated the phosphorylation of ERK1/2 and Elk-1 and upregulation of MMP-9, while pretreatment with telmisartan merely decreased the phosphorylation of Elk-1 and MMP-9 expression. These results suggested that IL-1β, IL-6, TGF-β, and MMP-9 participate in the pathophysiological process after CAS. Our new in vitro model mimics monocytes activated by stenting. MMP-9 expression could be regulated through ERK1/2/Elk-1 pathway, and the protective effects of telmisartan after stenting are partly attributed to its MMP-9 inhibition effects via suppression of Elk-1.
Collapse
|
35
|
Steinl DC, Kaufmann BA. Ultrasound imaging for risk assessment in atherosclerosis. Int J Mol Sci 2015; 16:9749-69. [PMID: 25938969 PMCID: PMC4463615 DOI: 10.3390/ijms16059749] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 04/09/2015] [Accepted: 04/09/2015] [Indexed: 01/28/2023] Open
Abstract
Atherosclerosis and its consequences like acute myocardial infarction or stroke are highly prevalent in western countries, and the incidence of atherosclerosis is rapidly rising in developing countries. Atherosclerosis is a disease that progresses silently over several decades before it results in the aforementioned clinical consequences. Therefore, there is a clinical need for imaging methods to detect the early stages of atherosclerosis and to better risk stratify patients. In this review, we will discuss how ultrasound imaging can contribute to the detection and risk stratification of atherosclerosis by (a) detecting advanced and early plaques; (b) evaluating the biomechanical consequences of atherosclerosis in the vessel wall;
Collapse
Affiliation(s)
- David C Steinl
- Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, Basel 4031, Switzerland.
| | - Beat A Kaufmann
- Division of Cardiology, University Hospital Basel, Petersgraben 4, Basel 4031, Switzerland.
| |
Collapse
|
36
|
Caveolin-1 regulates the anti-atherogenic properties of macrophages. Cell Tissue Res 2014; 358:821-31. [PMID: 25322709 DOI: 10.1007/s00441-014-2008-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 09/11/2014] [Indexed: 10/24/2022]
Abstract
Atherosclerosis is a complex disease initiated by the vascular accumulation of lipoproteins in the sub-endothelial space, followed by the infiltration of monocytes into the arterial intima. Caveolin-1 (Cav-1) plays an essential role in the regulation of cellular cholesterol metabolism and of various signaling pathways. In order to study specifically the role of macrophage Cav-1 in atherosclerosis, we used Cav-1 (-/-) Apoe (-/-) mice and transplanted them with bone marrow (BM) cells obtained from Cav-1 (+/+) Apoe (-/-) or Cav-1 (-/-) Apoe (-/-) mice and vice versa. We found that Cav-1 (+/+) mice harboring Cav-1 (-/-) BM-derived macrophages developed significantly larger lesions than Cav-1 (+/+) mice harboring Cav-1 (+/+) BM-derived macrophages. Cav-1 (-/-) macrophages were more susceptible to apoptosis and more prone to induce inflammation. The present study provides clear evidence that the absence of Cav-1 in macrophage is pro-atherogenic, whereas its absence in endothelial cells protects against atherosclerotic lesion formation. These findings demonstrate the cell-specific role of Cav-1 during the development of this disease.
Collapse
|
37
|
Kaynar AM, Yende S, Zhu L, Frederick DR, Chambers R, Burton CL, Carter M, Stolz DB, Agostini B, Gregory AD, Nagarajan S, Shapiro SD, Angus DC. Effects of intra-abdominal sepsis on atherosclerosis in mice. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2014; 18:469. [PMID: 25182529 PMCID: PMC4172909 DOI: 10.1186/s13054-014-0469-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 07/22/2014] [Indexed: 11/10/2022]
Abstract
Introduction Sepsis and other infections are associated with late cardiovascular events. Although persistent inflammation is implicated, a causal relationship has not been established. We tested whether sepsis causes vascular inflammation and accelerates atherosclerosis. Methods We performed prospective, randomized animal studies at a university research laboratory involving adult male ApoE-deficient (ApoE−/−) and young C57B/L6 wild-type (WT) mice. In the primary study conducted to determine whether sepsis accelerates atherosclerosis, we fed ApoE−/− mice (N = 46) an atherogenic diet for 4 months and then performed cecal ligation and puncture (CLP), followed by antibiotic therapy and fluid resuscitation or a sham operation. We followed mice for up to an additional 5 months and assessed atheroma in the descending aorta and root of the aorta. We also exposed 32 young WT mice to CLP or sham operation and followed them for 5 days to determine the effects of sepsis on vascular inflammation. Results ApoE−/− mice that underwent CLP had reduced activity during the first 14 days (38% reduction compared to sham; P < 0.001) and sustained weight loss compared to the sham-operated mice (−6% versus +9% change in weight after CLP or sham surgery to 5 months; P < 0.001). Despite their weight loss, CLP mice had increased atheroma (46% by 3 months and 41% increase in aortic surface area by 5 months; P = 0.03 and P = 0.004, respectively) with increased macrophage infiltration into atheroma as assessed by immunofluorescence microscopy (0.52 relative fluorescence units (rfu) versus 0.97 rfu; P = 0.04). At 5 months, peritoneal cultures were negative; however, CLP mice had elevated serum levels of interleukin 6 (IL-6) and IL-10 (each at P < 0.05). WT mice that underwent CLP had increased expression of intercellular adhesion molecule 1 in the aortic lumen versus sham at 24 hours (P = 0.01) that persisted at 120 hours (P = 0.006). Inflammatory and adhesion genes (tumor necrosis factor α, chemokine (C-C motif) ligand 2 and vascular cell adhesion molecule 1) and the adhesion assay, a functional measure of endothelial activation, were elevated at 72 hours and 120 hours in mice that underwent CLP versus sham-operations (all at P <0.05). Conclusions Using a combination of existing murine models for atherosclerosis and sepsis, we found that CLP, a model of intra-abdominal sepsis, accelerates atheroma development. Accelerated atheroma burden was associated with prolonged systemic, endothelial and intimal inflammation and was not explained by ongoing infection. These findings support observations in humans and demonstrate the feasibility of a long-term follow-up murine model of sepsis. Electronic supplementary material The online version of this article (doi:10.1186/s13054-014-0469-1) contains supplementary material, which is available to authorized users.
Collapse
|
38
|
Chang RCA, Ying W, Bazer FW, Zhou B. MicroRNAs Control Macrophage Formation and Activation: The Inflammatory Link between Obesity and Cardiovascular Diseases. Cells 2014; 3:702-712. [PMID: 25014161 PMCID: PMC4197627 DOI: 10.3390/cells3030702] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 06/27/2014] [Accepted: 07/01/2014] [Indexed: 12/16/2022] Open
Abstract
Activation and recruitment of resident macrophages in tissues in response to physiological stress are crucial regulatory processes in promoting the development of obesity-associated metabolic disorders and cardiovascular diseases. Recent studies have provided compelling evidence that microRNAs play important roles in modulating monocyte formation, macrophage maturation, infiltration into tissues and activation. Macrophage-dependent systemic physiological and tissue-specific responses also involve cell-cell interactions between macrophages and host tissue niche cell components, including other tissue-resident immune cell lineages, adipocytes, vascular smooth muscle and others. In this review, we highlight the roles of microRNAs in regulating the development and function of macrophages in the context of obesity, which could provide insights into the pathogenesis of obesity-related metabolic syndrome and cardiovascular diseases.
Collapse
Affiliation(s)
- Richard Cheng-An Chang
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - Wei Ying
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA
| | - Beiyan Zhou
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
39
|
Martorell J, Santomá P, Kolandaivelu K, Kolachalama VB, Melgar-Lesmes P, Molins JJ, Garcia L, Edelman ER, Balcells M. Extent of flow recirculation governs expression of atherosclerotic and thrombotic biomarkers in arterial bifurcations. Cardiovasc Res 2014; 103:37-46. [PMID: 24841070 PMCID: PMC4670884 DOI: 10.1093/cvr/cvu124] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 04/10/2014] [Accepted: 05/01/2014] [Indexed: 11/12/2022] Open
Abstract
AIMS Atherogenesis, evolution of plaque, and outcomes following endovascular intervention depend heavily on the unique vascular architecture of each individual. Patient-specific, multiscale models able to correlate changes in microscopic cellular responses with relevant macroscopic flow, and structural conditions may help understand the progression of occlusive arterial disease, providing insights into how to mitigate adverse responses in specific settings and individuals. METHODS AND RESULTS Vascular architectures mimicking coronary and carotid bifurcations were derived from clinical imaging and used to generate conjoint computational meshes for in silico analysis and biocompatible scaffolds for in vitro models. In parallel with three-dimensional flow simulations, geometrically realistic scaffolds were seeded with human smooth muscle cells (SMC) or endothelial cells and exposed to relevant, physiological flows. In vitro surrogates of endothelial health, atherosclerotic progression, and thrombosis were locally quantified and correlated best with an quantified extent of flow recirculation occurring within the bifurcation models. Oxidized low-density lipoprotein uptake, monocyte adhesion, and tissue factor expression locally rose up to three-fold, and phosphorylated endothelial nitric oxide synthase and Krüppel-like factor 2 decreased up to two-fold in recirculation areas. Isolated testing in straight-tube idealized constructs subject to static, oscillatory, and pulsatile conditions, indicative of different recirculant conditions corroborated these flow-mediated dependencies. CONCLUSIONS Flow drives variations in vascular reactivity and vascular beds. Endothelial health was preserved by arterial flow but jeopardized in regions of flow recirculation in a quasi-linear manner. Similarly, SMC exposed to flow were more thrombogenic in large recirculating regions. Health, thrombosis, and atherosclerosis biomarkers correlate with the extent of recirculation in vascular cells lining certain vascular geometries.
Collapse
Affiliation(s)
- Jordi Martorell
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA Department of Chemical Engineering, IQS School of Engineering, URL, Via Augusta 390, 08017 Barcelona, Spain
| | - Pablo Santomá
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA Department of Chemical Engineering, IQS School of Engineering, URL, Via Augusta 390, 08017 Barcelona, Spain
| | - Kumaran Kolandaivelu
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Vijaya B Kolachalama
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA Charles Stark Draper Laboratory, Cambridge, MA, USA
| | - Pedro Melgar-Lesmes
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - José J Molins
- Department of Chemical Engineering, IQS School of Engineering, URL, Via Augusta 390, 08017 Barcelona, Spain
| | - Lawrence Garcia
- Department of Interventional Cardiology and Vascular Medicine, St. Elizabeth's Medical Center, Boston, MA, USA
| | - Elazer R Edelman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA Cardiovascular Division, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Mercedes Balcells
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA Department of Biological Engineering, IQS School of Engineering, URL, Barcelona, Spain
| |
Collapse
|
40
|
Smolock EM, Burke RM, Wang C, Thomas T, Batchu SN, Qiu X, Zettel M, Fujiwara K, Berk BC, Korshunov VA. Intima modifier locus 2 controls endothelial cell activation and vascular permeability. Physiol Genomics 2014; 46:624-33. [PMID: 24986958 DOI: 10.1152/physiolgenomics.00048.2014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Carotid intima formation is a significant risk factor for cardiovascular disease. C3H/FeJ (C3H/F) and SJL/J (SJL) inbred mouse strains differ in susceptibility to immune and vascular traits. Using a congenic approach we demonstrated that the Intima modifier 2 (Im2) locus on chromosome 11 regulates leukocyte infiltration. We sought to determine whether inflammation was due to changes in circulating immune cells or activation of vascular wall cells in genetically pure Im2 (C3H/F.SJL.11.1) mice. Complete blood counts showed no differences in circulating monocytes between C3H/F and C3H/F.SJL.11.1 compared with SJL mice. Aortic vascular cell adhesion molecule-1 (VCAM-1) total protein levels were dramatically increased in SJL and C3H/F.SJL.11.1 compared with C3H/F mice. Immunostaining of aortic endothelial cells (EC) showed a significant increase in VCAM-1 expression in SJL and C3H/F.SJL.11.1 compared with C3H/F under steady flow conditions. Immunostaining of EC membranes revealed a significant decrease in EC size in SJL and C3H/F.SJL.11.1 vs. C3H/F in regions of disturbed flow. Vascular permeability was significantly higher in C3H/F.SJL.11.1 compared with C3H/F. Our results indicate that Im2 regulation of leukocyte infiltration is mediated by EC inflammation and permeability. RNA sequencing and pathway analyses comparing genes in the Im2 locus to C3H/F provide insight into candidate genes that regulate vascular wall inflammation and permeability highlighting important genetic mechanisms that control vascular intima in response to injury.
Collapse
Affiliation(s)
- Elaine M Smolock
- Department of Medicine and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Ryan M Burke
- Department of Medicine and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Chenjing Wang
- Department of Medicine and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York; Function Teaching and Research Section, Medical College of Northwest University for Nationalities, Lanzhou, China
| | - Tamlyn Thomas
- Department of Medicine and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Sri N Batchu
- Department of Medicine and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Xing Qiu
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, New York; and
| | - Martha Zettel
- Department of Medicine and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Keigi Fujiwara
- Department of Medicine and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Bradford C Berk
- Department of Medicine and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York
| | - Vyacheslav A Korshunov
- Department of Medicine and Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York; Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, New York;
| |
Collapse
|
41
|
Kee PH, Kim H, Huang S, Laing ST, Moody MR, Vela D, Klegerman ME, McPherson DD. Nitric oxide pretreatment enhances atheroma component highlighting in vivo with intercellular adhesion molecule-1-targeted echogenic liposomes. ULTRASOUND IN MEDICINE & BIOLOGY 2014; 40:1167-76. [PMID: 24613216 PMCID: PMC4011946 DOI: 10.1016/j.ultrasmedbio.2013.12.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 12/04/2013] [Accepted: 12/07/2013] [Indexed: 05/08/2023]
Abstract
We present an ultrasound technique for the detection of inflammatory changes in developing atheromas. We used contrast-enhanced ultrasound imaging with (i) microbubbles targeted to intercellular adhesion molecule-1 (ICAM-1), a molecule of adhesion involved in inflammatory processes in lesions of atheromas in New Zealand White rabbits, and (ii) pretreatment with nitric oxide-loaded microbubbles and ultrasound activation at the site of the endothelium to enhance the permeability of the arterial wall and the penetration of ICAM-1-targeted microbubbles. This procedure increases acoustic enhancement 1.2-fold. Pretreatment with nitric oxide-loaded echogenic liposomes and ultrasound activation can potentially facilitate the subsequent penetration of targeted echogenic liposomes into the arterial wall, thus allowing improved detection of inflammatory changes in developing atheromas.
Collapse
Affiliation(s)
- Patrick H Kee
- Division of Cardiology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA.
| | - Hyunggun Kim
- Division of Cardiology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Shaoling Huang
- Division of Cardiology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Susan T Laing
- Division of Cardiology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Melanie R Moody
- Division of Cardiology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Deborah Vela
- Cardiovascular Pathology, The Texas Heart Institute, Houston, Texas, USA
| | - Melvin E Klegerman
- Division of Cardiology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - David D McPherson
- Division of Cardiology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
42
|
Koo HJ, Park HJ, Byeon HE, Kwak JH, Um SH, Kwon ST, Rhee DK, Pyo S. Chinese yam extracts containing β-sitosterol and ethyl linoleate protect against atherosclerosis in apolipoprotein E-deficient mice and inhibit muscular expression of VCAM-1 in vitro. J Food Sci 2014; 79:H719-29. [PMID: 24689699 DOI: 10.1111/1750-3841.12401] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 01/11/2014] [Indexed: 12/31/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease, which is associated with increased expression of adhesion molecules and monocyte recruitment into the arterial wall. This study evaluated whether hexane extracts from the edible part (DB-H1) or bark region (DB-H2) of Dioscorea. batatas Decne have anti-atherosclerotic properties in vivo and in vitro experiments. We also identified bioactive components in the hexane extracts. Thirty-six apolipoprotein E (ApoE(-/-) ) mice and 12 control (C57BL/6J) mice were given a Western-type diet for 11 or 21 wk. To examine the effects of yam extracts on lesion development, ApoE(-/-) mice were orally administered DB-H1 or DB-H2 for the duration of the study (200 mg/kg b.w./day, 3 times per wk). Both DB-H1 and DB-H2 significantly reduced the total atherosclerotic lesion area in the aortic root. In addition, plasma concentrations of total cholesterol, oxidized-low-density lipoprotein, and c-reactive protein were decreased by administration of DB-H1 and DB-H2. Consistent with the in vivo observations, DB-H1 and DB-H2 inhibited tumor necrosis factor (TNF)-α-induced vascular cell adhesion molecule-1 expression and adhesion of THP-1 monocytes to TNF-α-activated vascular smooth muscle cells. It was also found that treatment with DB-H1 or DB-H2 resulted in the inhibition nitric oxide (NO) and reactive oxygen species production and iNOS expression in macrophages. Thus, DB-H1 and DB-H2 seem to influence atherosclerosis by affecting the production of inflammatory mediators in vivo. Our results suggest that yam extracts have the potential to be used in the prevention of atherosclerosis.
Collapse
Affiliation(s)
- Hyun Jung Koo
- Div. of Immunopharmacology, School of Pharmacy, Sungkyunkwan Univ, Suwon, Gyunggi-do, 440-746, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Serebruany VL, Malinin A, Eisert C, Ong S. AGI-1067, a novel vascular protectant, anti-inflammatory drug and mild antiplatelet agent for treatment of atherosclerosis. Expert Rev Cardiovasc Ther 2014; 5:635-41. [PMID: 17605642 DOI: 10.1586/14779072.5.4.635] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Oxidation-sensitive signals play an important role in platelet activation. AGI-1067 is a novel, phenolic, intra- and extracellular antioxidant that inhibits the expression of a number of proinflammatory genes involved in atherosclerosis. AGI-1067 is the metabolically stable monosuccinic acid ester of probucol, and a potent phenolic antioxidant representing a novel class of orally bioavailable compounds termed vascular protectants. AGI-1067 exhibits antioxidant activity equipotent to probucol. In addition, animal studies have demonstrated dual pharmacological activities of AGI-1067: the ability to block the expression of oxidation-sensitive inflammatory genes including genes that code for vascular cell adhesion molecule-1 and monocyte chemotactic protein-1. Importantly, AGI-1067 also exhibits mild antiplatelet properties inhibiting surface expression of various key platelet receptors, the formation of platelet monocyte microparticles and PAR-1 thrombin receptors. AGI-1067 is currently being tested in the late trials, and if proven to improve clinical outcomes (ARISE trial), the drug will ultimately be used in patients with different manifestations of atherosclerosis and atherothrombosis.
Collapse
Affiliation(s)
- Victor L Serebruany
- HeartDrug Research Laboratories, Osler Medical Center, 7600 Osler Drive, Suite 307, Towson, MD 21204, USA.
| | | | | | | |
Collapse
|
44
|
Pavlides S, Gutierrez-Pajares JL, Iturrieta J, Lisanti MP, Frank PG. Endothelial caveolin-1 plays a major role in the development of atherosclerosis. Cell Tissue Res 2014; 356:147-57. [PMID: 24390341 DOI: 10.1007/s00441-013-1767-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 11/08/2013] [Indexed: 12/19/2022]
Abstract
Clinical studies have established the important impact of atherosclerotic disease in Western societies. This disease is characterized by the accumulation of lipids and the migration of various cell types in the sub-endothelial space of blood vessels. As demonstrated by many studies, endothelial cells play an essential role in the development of this disease. The endothelium acts as a gatekeeper of blood vessel integrity and cardiovascular health status. For instance, the transfer of lipids via the transport of lipoproteins in the arterial intima is believed to be mediated by endothelial cells through a process termed transcytosis. In addition, lipoproteins that accumulate in the sub-endothelial space may also be modified, in a process that can direct the activation of endothelial cells. These steps are essential for the initiation of an atherosclerotic plaque and may be mediated, at least in part, by caveolae and their associated protein caveolin-1. In the present study, we evaluate the role of caveolin-1/caveolae in the regulation of these two steps in endothelial cells. Our data clearly demonstrate that caveolin-1 is involved in the regulation of lipoprotein transcytosis across endothelial cells and in the regulation of vascular inflammation.
Collapse
Affiliation(s)
- Stephanos Pavlides
- Manchester Breast Centre & Breakthrough Breast Cancer Research Unit; Paterson Institute for Cancer Research; Institute of Cancer Sciences; Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | | | | | | | | |
Collapse
|
45
|
Schmitz B, Vischer P, Brand E, Schmidt-Petersen K, Korb-Pap A, Guske K, Nedele J, Schelleckes M, Hillen J, Rötrige A, Simmet T, Paul M, Cambien F, Brand SM. Increased monocyte adhesion by endothelial expression of VCAM-1 missense variation in vitro. Atherosclerosis 2013; 230:185-90. [DOI: 10.1016/j.atherosclerosis.2013.07.039] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Revised: 07/02/2013] [Accepted: 07/17/2013] [Indexed: 01/21/2023]
|
46
|
Yang BL, Wu S, Wu X, Li MB, Zhu W, Guan Y, Liu LH, Luo MY, Cai WJ, Schaper J, Schaper W. Effect of shunting of collateral flow into the venous system on arteriogenesis and angiogenesis in rabbit hind limb. Acta Histochem Cytochem 2013; 46:1-10. [PMID: 23554534 PMCID: PMC3596601 DOI: 10.1267/ahc.12025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2012] [Accepted: 10/05/2012] [Indexed: 01/31/2023] Open
Abstract
The aim of this study was to characterize the vascular remodeling in the external iliac artery (EIA) and the lower leg muscles in a rabbit shunt model created between the distal stump of the occluded femoral artery and the accompanying vein. Histology and immunoconfocal microscopy were used in this study. We found that: 1) both endothelial nitric oxide synthase (eNOS) and phosphorylated eNOS (P-eNOS) proteins were significantly increased in the shunt-side EIA; 2) matrix metalloproteinase-2 (MMP-2) expression was 5.5 times in shunt side EIA over that in normal EIA; 3) intercellular adhension molecule-1 (ICAM-1) expression was strongly induced in endothelial cells (EC) and vascular adhension molecule-1 (VCAM-1) expression was significantly increased in both EC and the adventitia of the shunt-side EIA; 4) augmentation of cell proliferation and extracellular proteolysis by macrophage infiltration was observed in shunt-side EIA; 5) cell proliferation was active in shunt side EIA, but quiet in shunt side lower leg’s arterial vessels; 6) capillary density in shunt side lower leg muscles was 2 times over that in normal side. In conclusion, our data demonstrate the paradigm that the power of shear stress takes the reins in arteriogenesis, whereas ischemia in angiogenesis, but not in arteriogenesis.
Collapse
Affiliation(s)
- Bao-lin Yang
- Department of Histology & Embryology, School of Basic Medicine, Central South University
- Department of Human Anatomy, School of Basic Medicine, Nanchang University
| | - Song Wu
- Department of Orthopedics, The 3rd Xiangya Hospital, Central South University
| | - Xiaoqiong Wu
- Department of Anatomy & Neurobiology, School of Basic Medicine, Central South University
| | - Ming Bo Li
- Department of Anatomy & Neurobiology, School of Basic Medicine, Central South University
| | - Wu Zhu
- Department of Histology & Embryology, School of Basic Medicine, Central South University
| | - Yinglu Guan
- Department of Histology & Embryology, School of Basic Medicine, Central South University
| | - Li-Hua Liu
- Department of Histology & Embryology, School of Basic Medicine, Central South University
| | - Ming-ying Luo
- Department of Histology & Embryology, School of Basic Medicine, Central South University
| | - Wei-Jun Cai
- Department of Histology & Embryology, School of Basic Medicine, Central South University
| | - Jutta Schaper
- Max-Planck-Institute for Heart and Lung Research, Arteriogenesis Research Group
| | - Wolfgang Schaper
- Max-Planck-Institute for Heart and Lung Research, Arteriogenesis Research Group
| |
Collapse
|
47
|
Cardilo-Reis L, Gruber S, Schreier SM, Drechsler M, Papac-Milicevic N, Weber C, Wagner O, Stangl H, Soehnlein O, Binder CJ. Interleukin-13 protects from atherosclerosis and modulates plaque composition by skewing the macrophage phenotype. EMBO Mol Med 2013; 4:1072-86. [PMID: 23027612 PMCID: PMC3491837 DOI: 10.1002/emmm.201201374] [Citation(s) in RCA: 208] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Atherosclerotic lesions are characterized by the accumulation of oxidized LDL (OxLDL) and the infiltration of macrophages and T cells. Cytokine expression in the microenvironment of evolving lesions can profoundly contribute to plaque development. While the pro-atherogenic effect of T helper (Th) 1 cytokines, such as IFN-γ, is well established, the role of Th2 cytokines is less clear. Therefore, we characterized the role of the Th2 cytokine interleukin (IL)-13 in murine atherosclerosis. Here, we report that IL-13 administration favourably modulated the morphology of already established atherosclerotic lesions by increasing lesional collagen content and reducing vascular cell adhesion molecule-1 (VCAM-1)-dependent monocyte recruitment, resulting in decreased plaque macrophage content. This was accompanied by the induction of alternatively activated (M2) macrophages, which exhibited increased clearance of OxLDL compared to IFN-γ-activated (M1) macrophages in vitro. Importantly, deficiency of IL-13 results in accelerated atherosclerosis in LDLR−/− mice without affecting plasma cholesterol levels. Thus, IL-13 protects from atherosclerosis and promotes a favourable plaque morphology, in part through the induction of alternatively activated macrophages.
Collapse
Affiliation(s)
- Larissa Cardilo-Reis
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Johnson LA, Kim HS, Knudson MJ, Nipp CT, Yi X, Maeda N. Diabetic atherosclerosis in APOE*4 mice: synergy between lipoprotein metabolism and vascular inflammation. J Lipid Res 2013; 54:386-96. [PMID: 23204275 PMCID: PMC3588868 DOI: 10.1194/jlr.m031435] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2012] [Revised: 11/29/2012] [Indexed: 11/20/2022] Open
Abstract
Diabetes is a major risk factor for cardiovascular disease. To examine how diabetes interacts with a mildly compromised lipid metabolism, we introduced the diabetogenic Ins2(C96Y/+) (Akita) mutation into mice expressing human apoE4 (E4) combined with either an overexpressing human LDL receptor gene (hLDLR) or the wild-type mouse gene. The hLDLR allele caused 2-fold reductions in plasma HDL-cholesterol, plasma apoA1, and hepatic triglyceride secretion. Diabetes increased plasma total cholesterol 1.3-fold and increased apoB48 secretion 3-fold, while reducing triglyceride secretion 2-fold. Consequently, diabetic E4 mice with hLDLR secrete increased numbers of small, cholesterol-enriched, apoB48-containing VLDL, although they have near normal plasma cholesterol (<120 mg/dl). Small foam cell lesions were present in the aortic roots of all diabetic E4 mice with hLDLR that we analyzed at six months of age. None were present in nondiabetic mice or in diabetic mice without hLDLR. Aortic expression of genes affecting leukocyte recruitment and adhesion was enhanced by diabetes. ApoA1 levels, but not diabetes, were strongly correlated with the ability of plasma to efflux cholesterol from macrophages. We conclude that the diabetes-induced proinflammatory changes in the vasculature and the hLDLR-mediated cholesterol accumulation in macrophages synergistically trigger atherosclerosis in mice with human apoE4, although neither alone is sufficient.
Collapse
Affiliation(s)
- Lance A. Johnson
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Hyung-Suk Kim
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Melissa J. Knudson
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - C. Taylor Nipp
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Xianwen Yi
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Nobuyo Maeda
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
49
|
McAteer MA, Choudhury RP. Targeted molecular imaging of vascular inflammation in cardiovascular disease using nano- and micro-sized agents. Vascul Pharmacol 2013; 58:31-8. [DOI: 10.1016/j.vph.2012.10.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 10/19/2012] [Indexed: 01/15/2023]
|
50
|
De Meyer I, Martinet W, De Meyer GRY. Therapeutic strategies to deplete macrophages in atherosclerotic plaques. Br J Clin Pharmacol 2012; 74:246-63. [PMID: 22309283 DOI: 10.1111/j.1365-2125.2012.04211.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Macrophages can be found in all stages of atherosclerosis and are major contributors of atherosclerotic plaque development, progression and destabilization. Continuous recruitment of monocytes drives this chronic inflammatory disease, which can be intervened by several strategies: reducing the inflammatory stimulus by lowering circulating lipids and promoting cholesterol efflux from plaque, direct and indirect targeting of adhesion molecules and chemokines involved in monocyte adhesion and transmigration and inducing macrophage death in atherosclerotic plaques in combination with anti-inflammatory drugs. This review discusses the outlined strategies to deplete macrophages from atherosclerotic plaques to promote plaque stabilization.
Collapse
Affiliation(s)
- Inge De Meyer
- Division of Physiopharmacology, University of Antwerp, Antwerp, Belgium.
| | | | | |
Collapse
|