251
|
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
|
252
|
Ha SJ, Lee J, Song KM, Kim YH, Lee NH, Kim YE, Jung SK. Ultrasonicated Lespedeza cuneata extract prevents TNF-α-induced early atherosclerosis in vitro and in vivo. Food Funct 2018; 9:2090-2101. [DOI: 10.1039/c7fo01666b] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This study evaluated the use of ultrasonication to extract Lespedeza cuneata as a potential nutraceutical for preventing vascular inflammation.
Collapse
Affiliation(s)
- Su Jeong Ha
- Division of Functional Food Research
- Korea Food Research Institute
- Jeollabuk-do 55365
- Republic of Korea
- Department of Agricultural Biotechnology
| | - Jangho Lee
- Division of Functional Food Research
- Korea Food Research Institute
- Jeollabuk-do 55365
- Republic of Korea
- Department of Food Biotechnology
| | - Kyung-Mo Song
- Division of Functional Food Research
- Korea Food Research Institute
- Jeollabuk-do 55365
- Republic of Korea
| | - Young Ho Kim
- Division of Functional Food Research
- Korea Food Research Institute
- Jeollabuk-do 55365
- Republic of Korea
| | - Nam Hyouck Lee
- Division of Functional Food Research
- Korea Food Research Institute
- Jeollabuk-do 55365
- Republic of Korea
| | - Young-Eon Kim
- Division of Functional Food Research
- Korea Food Research Institute
- Jeollabuk-do 55365
- Republic of Korea
| | - Sung Keun Jung
- Division of Functional Food Research
- Korea Food Research Institute
- Jeollabuk-do 55365
- Republic of Korea
- School of Food Science and Biotechnology
| |
Collapse
|
253
|
Zhu L, Shi J, Luu TN, Neuman JC, Trefts E, Yu S, Palmisano BT, Wasserman DH, Linton MF, Stafford JM. Hepatocyte estrogen receptor alpha mediates estrogen action to promote reverse cholesterol transport during Western-type diet feeding. Mol Metab 2017; 8:106-116. [PMID: 29331506 PMCID: PMC5985047 DOI: 10.1016/j.molmet.2017.12.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 12/16/2017] [Accepted: 12/23/2017] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Hepatocyte deletion of estrogen receptor alpha (LKO-ERα) worsens fatty liver, dyslipidemia, and insulin resistance in high-fat diet fed female mice. However, whether or not hepatocyte ERα regulates reverse cholesterol transport (RCT) in mice has not yet been reported. METHODS AND RESULTS Using LKO-ERα mice and wild-type (WT) littermates fed a Western-type diet, we found that deletion of hepatocyte ERα impaired in vivo RCT measured by the removal of 3H-cholesterol from macrophages to the liver, and subsequently to feces, in female mice but not in male mice. Deletion of hepatocyte ERα decreased the capacity of isolated HDL to efflux cholesterol from macrophages and reduced the ability of isolated hepatocytes to accept cholesterol from HDL ex vivo in both sexes. However, only in female mice, LKO-ERα increased serum cholesterol levels and increased HDL particle sizes. Deletion of hepatocyte ERα increased adiposity and worsened insulin resistance to a greater degree in female than male mice. All of the changes lead to a 5.6-fold increase in the size of early atherosclerotic lesions in female LKO-ERα mice compared to WT controls. CONCLUSIONS Estrogen signaling through hepatocyte ERα plays an important role in RCT and is protective against lipid retention in the artery wall during early stages of atherosclerosis in female mice fed a Western-type diet.
Collapse
Affiliation(s)
- Lin Zhu
- VA Tennessee Valley Healthcare System, USA; Division of Diabetes, Endocrinology, & Metabolism, USA
| | - Jeanne Shi
- Division of Diabetes, Endocrinology, & Metabolism, USA; Trinity College of Arts and Sciences, Duke University, USA
| | - Thao N Luu
- Division of Diabetes, Endocrinology, & Metabolism, USA
| | | | - Elijah Trefts
- Department of Molecular, Physiology and Biophysics, Vanderbilt University, USA
| | - Sophia Yu
- Division of Diabetes, Endocrinology, & Metabolism, USA
| | - Brian T Palmisano
- Department of Molecular, Physiology and Biophysics, Vanderbilt University, USA
| | - David H Wasserman
- Department of Molecular, Physiology and Biophysics, Vanderbilt University, USA
| | - MacRae F Linton
- Division of Cardiovascular Medicine, Vanderbilt University School of Medicine, USA
| | - John M Stafford
- VA Tennessee Valley Healthcare System, USA; Department of Molecular, Physiology and Biophysics, Vanderbilt University, USA; Division of Diabetes, Endocrinology, & Metabolism, USA.
| |
Collapse
|
254
|
Rinne P, Kadiri JJ, Velasco-Delgado M, Nuutinen S, Viitala M, Hollmén M, Rami M, Savontaus E, Steffens S. Melanocortin 1 Receptor Deficiency Promotes Atherosclerosis in Apolipoprotein E -/- Mice. Arterioscler Thromb Vasc Biol 2017; 38:313-323. [PMID: 29284608 PMCID: PMC5779319 DOI: 10.1161/atvbaha.117.310418] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 12/18/2017] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The MC1-R (melanocortin 1 receptor) is expressed by monocytes and macrophages where it mediates anti-inflammatory actions. MC1-R also protects against macrophage foam cell formation primarily by promoting cholesterol efflux through the ABCA1 (ATP-binding cassette transporter subfamily A member 1) and ABCG1 (ATP-binding cassette transporter subfamily G member 1). In this study, we aimed to investigate whether global deficiency in MC1-R signaling affects the development of atherosclerosis. APPROACH AND RESULTS Apoe-/- (apolipoprotein E deficient) mice were crossed with recessive yellow (Mc1re/e) mice carrying dysfunctional MC1-R and fed a high-fat diet to induce atherosclerosis. Apoe-/- Mc1re/e mice developed significantly larger atherosclerotic lesions in the aortic sinus and in the whole aorta compared with Apoe-/- controls. In terms of plaque composition, MC1-R deficiency was associated with less collagen and smooth muscle cells and increased necrotic core, indicative of more vulnerable lesions. These changes were accompanied by reduced Abca1 and Abcg1 expression in the aorta. Furthermore, Apoe-/- Mc1re/e mice showed a defect in bile acid metabolism that aggravated high-fat diet-induced hypercholesterolemia and hepatic lipid accumulation. Flow cytometric analysis of leukocyte profile revealed that dysfunctional MC1-R enhanced arterial accumulation of classical Ly6Chigh monocytes and macrophages, effects that were evident in mice fed a normal chow diet but not under high-fat diet conditions. In support of enhanced arterial recruitment of Ly6Chigh monocytes, these cells had increased expression of L-selectin and P-selectin glycoprotein ligand 1. CONCLUSIONS The present study highlights the importance of MC1-R in the development of atherosclerosis. Deficiency in MC1-R signaling exacerbates atherosclerosis by disturbing cholesterol handling and by increasing arterial monocyte accumulation.
Collapse
Affiliation(s)
- Petteri Rinne
- From the Institute of Biomedicine, Research Center for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Finland (P.R., J.J.K., M.V.-D., S.N., E.S.); Medicity Research Laboratory Turku, University of Turku, Finland (M.V., M.H.); Unit of Clinical Pharmacology, Turku University Hospital, Finland (E.S.); Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU), Munich, Germany (P.R., M.R., S.S.); and German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Germany (S.S.).
| | - James J Kadiri
- From the Institute of Biomedicine, Research Center for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Finland (P.R., J.J.K., M.V.-D., S.N., E.S.); Medicity Research Laboratory Turku, University of Turku, Finland (M.V., M.H.); Unit of Clinical Pharmacology, Turku University Hospital, Finland (E.S.); Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU), Munich, Germany (P.R., M.R., S.S.); and German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Germany (S.S.)
| | - Mauricio Velasco-Delgado
- From the Institute of Biomedicine, Research Center for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Finland (P.R., J.J.K., M.V.-D., S.N., E.S.); Medicity Research Laboratory Turku, University of Turku, Finland (M.V., M.H.); Unit of Clinical Pharmacology, Turku University Hospital, Finland (E.S.); Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU), Munich, Germany (P.R., M.R., S.S.); and German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Germany (S.S.)
| | - Salla Nuutinen
- From the Institute of Biomedicine, Research Center for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Finland (P.R., J.J.K., M.V.-D., S.N., E.S.); Medicity Research Laboratory Turku, University of Turku, Finland (M.V., M.H.); Unit of Clinical Pharmacology, Turku University Hospital, Finland (E.S.); Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU), Munich, Germany (P.R., M.R., S.S.); and German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Germany (S.S.)
| | - Miro Viitala
- From the Institute of Biomedicine, Research Center for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Finland (P.R., J.J.K., M.V.-D., S.N., E.S.); Medicity Research Laboratory Turku, University of Turku, Finland (M.V., M.H.); Unit of Clinical Pharmacology, Turku University Hospital, Finland (E.S.); Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU), Munich, Germany (P.R., M.R., S.S.); and German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Germany (S.S.)
| | - Maija Hollmén
- From the Institute of Biomedicine, Research Center for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Finland (P.R., J.J.K., M.V.-D., S.N., E.S.); Medicity Research Laboratory Turku, University of Turku, Finland (M.V., M.H.); Unit of Clinical Pharmacology, Turku University Hospital, Finland (E.S.); Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU), Munich, Germany (P.R., M.R., S.S.); and German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Germany (S.S.)
| | - Martina Rami
- From the Institute of Biomedicine, Research Center for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Finland (P.R., J.J.K., M.V.-D., S.N., E.S.); Medicity Research Laboratory Turku, University of Turku, Finland (M.V., M.H.); Unit of Clinical Pharmacology, Turku University Hospital, Finland (E.S.); Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU), Munich, Germany (P.R., M.R., S.S.); and German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Germany (S.S.)
| | - Eriika Savontaus
- From the Institute of Biomedicine, Research Center for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Finland (P.R., J.J.K., M.V.-D., S.N., E.S.); Medicity Research Laboratory Turku, University of Turku, Finland (M.V., M.H.); Unit of Clinical Pharmacology, Turku University Hospital, Finland (E.S.); Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU), Munich, Germany (P.R., M.R., S.S.); and German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Germany (S.S.)
| | - Sabine Steffens
- From the Institute of Biomedicine, Research Center for Integrative Physiology and Pharmacology, Turku Center for Disease Modeling, University of Turku, Finland (P.R., J.J.K., M.V.-D., S.N., E.S.); Medicity Research Laboratory Turku, University of Turku, Finland (M.V., M.H.); Unit of Clinical Pharmacology, Turku University Hospital, Finland (E.S.); Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU), Munich, Germany (P.R., M.R., S.S.); and German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Germany (S.S.)
| |
Collapse
|
255
|
Buscher K, Marcovecchio P, Hedrick CC, Ley K. Patrolling Mechanics of Non-Classical Monocytes in Vascular Inflammation. Front Cardiovasc Med 2017; 4:80. [PMID: 29312957 PMCID: PMC5742122 DOI: 10.3389/fcvm.2017.00080] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/23/2017] [Indexed: 12/13/2022] Open
Abstract
Non-classical monocytes have emerged as the preeminent vascular housekeepers. Continuous intravascular screening is enabled by slow patrolling on the endothelium and allows a rapid response to local perturbations. Intravital imaging has been crucial to elucidate the molecular mechanisms and migratory phenotype of patrolling. In this review, we discuss technical requirements of intravital microscopy such as imaging modalities, labeling strategies, and data analysis. We further focus on patrolling kinetics and adhesion receptors in different organs and vascular beds including arteries during homeostasis and vascular inflammation and define pertinent questions in the field.
Collapse
Affiliation(s)
- Konrad Buscher
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States.,Department of Internal Medicine, Nephrology and Rheumatology, University Hospital Münster, Münster, Germany
| | - Paola Marcovecchio
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| | - Catherine C Hedrick
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| |
Collapse
|
256
|
Sager HB, Dutta P, Dahlman JE, Hulsmans M, Courties G, Sun Y, Heidt T, Vinegoni C, Borodovsky A, Fitzgerald K, Wojtkiewicz GR, Iwamoto Y, Tricot B, Khan OF, Kauffman KJ, Xing Y, Shaw TE, Libby P, Langer R, Weissleder R, Swirski FK, Anderson DG, Nahrendorf M. RNAi targeting multiple cell adhesion molecules reduces immune cell recruitment and vascular inflammation after myocardial infarction. Sci Transl Med 2017; 8:342ra80. [PMID: 27280687 DOI: 10.1126/scitranslmed.aaf1435] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 05/17/2016] [Indexed: 12/22/2022]
Abstract
Myocardial infarction (MI) leads to a systemic surge of vascular inflammation in mice and humans, resulting in secondary ischemic complications and high mortality. We show that, in ApoE(-/-) mice with coronary ligation, increased sympathetic tone up-regulates not only hematopoietic leukocyte production but also plaque endothelial expression of adhesion molecules. To counteract the resulting arterial leukocyte recruitment, we developed nanoparticle-based RNA interference (RNAi) that effectively silences five key adhesion molecules. Simultaneously encapsulating small interfering RNA (siRNA)-targeting intercellular cell adhesion molecules 1 and 2 (Icam1 and Icam2), vascular cell adhesion molecule 1 (Vcam1), and E- and P-selectins (Sele and Selp) into polymeric endothelial-avid nanoparticles reduced post-MI neutrophil and monocyte recruitment into atherosclerotic lesions and decreased matrix-degrading plaque protease activity. Five-gene combination RNAi also curtailed leukocyte recruitment to ischemic myocardium. Therefore, targeted multigene silencing may prevent complications after acute MI.
Collapse
Affiliation(s)
- Hendrik B Sager
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Partha Dutta
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - James E Dahlman
- Harvard-Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, Cambridge, MA 02139, USA.,David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA.,Institute for Medical Engineering and Science, MIT, Cambridge, MA 02139, USA
| | - Maarten Hulsmans
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Gabriel Courties
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Yuan Sun
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Timo Heidt
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Claudio Vinegoni
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | | | - Gregory R Wojtkiewicz
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Yoshiko Iwamoto
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Benoit Tricot
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Omar F Khan
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Kevin J Kauffman
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA.,Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA
| | - Yiping Xing
- Harvard-Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, Cambridge, MA 02139, USA.,David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Taylor E Shaw
- Harvard-Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, Cambridge, MA 02139, USA.,David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Peter Libby
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Robert Langer
- Harvard-Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, Cambridge, MA 02139, USA.,David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA.,Institute for Medical Engineering and Science, MIT, Cambridge, MA 02139, USA.,Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.,Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Filip K Swirski
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Daniel G Anderson
- Harvard-Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, Cambridge, MA 02139, USA.,David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA.,Institute for Medical Engineering and Science, MIT, Cambridge, MA 02139, USA.,Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.,Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
257
|
Ungprasert P, Matteson EL, Crowson CS. Increased Risk of Multimorbidity in Patients With Sarcoidosis: A Population-Based Cohort Study 1976 to 2013. Mayo Clin Proc 2017; 92:1791-1799. [PMID: 29108842 PMCID: PMC5763921 DOI: 10.1016/j.mayocp.2017.09.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 09/01/2017] [Accepted: 09/25/2017] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To evaluate the risk and pattern of multimorbidity in patients with sarcoidosis. PATIENTS AND METHODS A cohort of all residents of Olmsted County, Minnesota, first diagnosed with sarcoidosis between January 1, 1976, and December 31, 2013, was identified through the medical record linkage system of the Rochester Epidemiology Project. Diagnosis was verified by individual medical record review. A cohort of sex- and age-matched comparators without sarcoidosis was assembled from the same population. Data on 18 chronic conditions recommended by the US Department of Health and Human Services for both cases and comparators were retrieved and compared. RESULTS The prevalence of multimorbidity (ie, the presence of ≥2 chronic conditions) was similar between the 2 groups: 111 of 345 cases (32.2%) and 110 of 345 comparators (31.9%) (P=.99). After the index date, 156 cases (43.8%) and 142 comparators (41.2%) developed multimorbidity, corresponding to a hazard ratio of 1.60 (95% CI, 1.27-2.01; P<.001). The cumulative incidence of the presence of ≥3, 4, and 5 chronic conditions was also consistently significantly higher in cases than in comparators (P value=.01, .004 and .002, respectively). Analysis by specific type of chronic condition revealed a significantly higher cumulative incidence of coronary artery disease, congestive heart failure, arrhythmia, stroke or transient ischemic attack, arthritis, depression, diabetes, and major osteoporotic fracture. CONCLUSION In this population, patients with sarcoidosis had a significantly higher risk of developing multimorbidity than did sex- and age-matched individuals without sarcoidosis.
Collapse
Affiliation(s)
- Patompong Ungprasert
- Division of Rheumatology, Mayo Clinic, Rochester, MN; Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| | - Eric L Matteson
- Division of Rheumatology, Mayo Clinic, Rochester, MN; Division of Epidemiology, Mayo Clinic, Rochester, MN
| | - Cynthia S Crowson
- Division of Rheumatology, Mayo Clinic, Rochester, MN; Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN
| |
Collapse
|
258
|
Asare Y, Shagdarsuren E, Schmid J, Tilstam P, Grommes J, El Bounkari O, Schütz A, Weber C, de Winther M, Noels H, Bernhagen J. Endothelial CSN5 impairs NF-κB activation and monocyte adhesion to endothelial cells and is highly expressed in human atherosclerotic lesions. Thromb Haemost 2017; 110:141-52. [DOI: 10.1160/th13-02-0155] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 04/05/2013] [Indexed: 01/07/2023]
Abstract
SummaryThe COP9 signalosome (CSN), a multifunctional protein complex involved in the regulation of cullin-RING-E3 ubiquitin ligases (CRLs), has emerged as a regulator of NF-κB signalling. As NF-κB drives the expression of pro-inflammatory and pro-atherosclerotic genes, we probed the yet unknown role of the CSN, in particular CSN5, on NF-KB-mediated atherogenic responses in endothelial cells. Co-immunoprecipitation in human umbilical vein endothelial cells (HUVECs) revealed the presence of a super-complex between IKK and CSN, which dissociates upon TNF-α stimulation. Furthermore, CSN5 silencing enhanced TNF-α-induced IKB-α degradation and NF-κB activity in luci-ferase reporter assays. This was paralleled by an increased NF-KB-driven upregulation of atherogenic chemokines and adhesion molecules, as measured by qPCR and flow cytometry, and translated into an enhanced arrest of THP-1 monocytes on TNF-α-stimulated, CSN5-depleted HUVECs. Reverse effects on NF-κB activity and THP-1 arrest were seen upon CSN5 overexpression. Finally, double-immunostaining confirmed the expression of CSN subunits in the endothelium of human atherosclerotic lesions, and revealed an increased expression of CSN5 which correlated with atheroprogression. In conclusion, endothelial CSN5 attenuates NF-KB-dependent pro-inflammatory gene expression and monocyte arrest on stimulated endothelial cells in vitro, suggesting that CSN5 might serve as a negative regulator of atherogenesis.Note: The review process for this manuscript was fully handled by G. Y. H. Lip, Editor in Chief.
Collapse
|
259
|
Chinese Herbal Cardiotonic Pill Stabilizes Vulnerable Plaques in Rabbits by Decreasing the Expression of Adhesion Molecules. J Cardiovasc Pharmacol 2017; 68:215-22. [PMID: 27110743 PMCID: PMC5049970 DOI: 10.1097/fjc.0000000000000403] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The cardiotonic pill (CP), consisting of a mixture of Radix Salviae Miltiorrhizae, Radix Notoginseng, and Borneolum Syntheticum, has been widely used in the prevention and treatment of cardiovascular disease. Adhesion molecules, including intercellular cell adhesion molecule-1 and vascular cell adhesion molecule-1, are involved in the development of vulnerable plaque. We investigated the effect of the CP in a rabbit model of vulnerable plaque established by local transfection with p53 gene. Compared with the control group, rabbits with vulnerable plaque showed a significantly lower intima-media thickness and plaque burden after CP treatment for 12 weeks. Moreover, the reduction in rate of plaque rupture and vulnerability index was similar. On enzyme-linked immunosorbent assay, real-time polymerase chain reaction, and immunohistochemistry analysis, the expression of intercellular cell adhesion molecule-1 and vascular cell adhesion molecule-1 was inhibited with CP treatment. CP treatment could postpone atherosclerotic plaque development and stabilize vulnerable plaque by inhibiting the expression of adhesion molecules in treatment of cardiovascular disease.
Collapse
|
260
|
Döring Y, Megens R, Soehnlein O, Drechsler M. Neutrophilic granulocytes – promiscuous accelerators of atherosclerosis. Thromb Haemost 2017; 106:839-48. [DOI: 10.1160/th11-07-0501] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Accepted: 09/28/2011] [Indexed: 12/23/2022]
Abstract
SummaryNeutrophils, as part of the innate immune system, are classically described to be main actors during the onset of inflammation enforcing rapid neutralisation and clearance of pathogens. Besides their wellstudied role in acute inflammatory processes, recent advances strongly indicate a so far underappreciated importance of neutrophils in initiation and development of atherosclerosis. This review focuses on current findings on the role of neutrophils in atherosclerosis. As pro-inflammatory mechanisms of neutrophils have primarily been studied in the microvascular environment; we here aim at translating these into the context of macrovascular inflammation in atherosclerosis. Since much of the pro-inflammatory activities of neutrophils stem from instructing neighbouring cell types, we highlight the promiscuous interplay between neutrophils and platelets, monocytes, T lymphocytes, and dendritic cells and its possible relevance to atherosclerosis.
Collapse
|
261
|
Park K, Li Q, Evcimen ND, Rask-Madsen C, Maeda Y, Maddaloni E, Yokomizo H, Shinjo T, St-Louis R, Fu J, Gordin D, Khamaisi M, Pober D, Keenan H, King GL. Exogenous Insulin Infusion Can Decrease Atherosclerosis in Diabetic Rodents by Improving Lipids, Inflammation, and Endothelial Function. Arterioscler Thromb Vasc Biol 2017; 38:92-101. [PMID: 29162603 DOI: 10.1161/atvbaha.117.310291] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 11/09/2017] [Indexed: 01/14/2023]
Abstract
OBJECTIVE The objective of this study is to evaluate whether exogenously induced hyperinsulinemia may increase the development of atherosclerosis. APPROACH AND RESULTS Hyperinsulinemia, induced by exogenous insulin implantation in high-fat fed (60% fat HFD) apolipoprotein E-deficient mice (ApoE-/-) mice, exhibited insulin resistance, hyperglycemia, and hyperinsulinemia. Atherosclerosis was measured by the accumulation of fat, macrophage, and extracellular matrix in the aorta. After 8 weeks on HFD, ApoE-/- mice were subcutaneously implanted with control (sham) or insulin pellet, and phlorizin, a sodium glucose cotransporters inhibitor (1/2)inhibitor, for additional 8 weeks. Intraperitoneal glucose tolerance test showed that plasma glucose levels were lower and insulin and IGF-1 (insulin-like growth factor-1) levels were 5.3- and 3.3-fold higher, respectively, in insulin-implanted compared with sham-treated ApoE-/- mice. Plasma triglyceride, cholesterol, and lipoprotein levels were decreased in mice with insulin implant, in parallel with increased lipoprotein lipase activities. Atherosclerotic plaque by en face and complexity staining showed significant reductions of fat deposits and expressions of vascular adhesion molecule-1, tumor necrosis factor-α, interleukin 6, and macrophages in arterial wall while exhibiting increased activation of pAKT and endothelial nitric oxide synthase (P<0.05) comparing insulin-implanted versus sham HFD ApoE-/- mice. No differences were observed in atherosclerotic plaques between phlorizin-treated and sham HFD ApoE-/- mice, except phlorizin significantly lowered plasma glucose and glycated hemoglobin levels while increased glucosuria. Endothelial function was improved only by insulin treatment through endothelial nitric oxide synthase/nitric oxide activations and reduced proinflammatory (M1) and increased anti-inflammatory (M2) macrophages, which were inhibited by endothelial nitric oxide synthase inhibitor. CONCLUSIONS Exogenous insulin decreased atherosclerosis by lowering inflammatory cytokines, macrophages, and plasma lipids in HFD-induced hyperlipidemia, insulin resistant and mildly diabetic ApoE-/- mice.
Collapse
Affiliation(s)
- Kyoungmin Park
- From the Dianne Nunnally Hoppes Laboratory, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Qian Li
- From the Dianne Nunnally Hoppes Laboratory, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Net Daş Evcimen
- From the Dianne Nunnally Hoppes Laboratory, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Christian Rask-Madsen
- From the Dianne Nunnally Hoppes Laboratory, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Yasutaka Maeda
- From the Dianne Nunnally Hoppes Laboratory, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Ernesto Maddaloni
- From the Dianne Nunnally Hoppes Laboratory, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Hisashi Yokomizo
- From the Dianne Nunnally Hoppes Laboratory, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Takanori Shinjo
- From the Dianne Nunnally Hoppes Laboratory, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Ronald St-Louis
- From the Dianne Nunnally Hoppes Laboratory, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Jialin Fu
- From the Dianne Nunnally Hoppes Laboratory, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Daniel Gordin
- From the Dianne Nunnally Hoppes Laboratory, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Mogher Khamaisi
- From the Dianne Nunnally Hoppes Laboratory, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - David Pober
- From the Dianne Nunnally Hoppes Laboratory, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Hillary Keenan
- From the Dianne Nunnally Hoppes Laboratory, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - George L King
- From the Dianne Nunnally Hoppes Laboratory, Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA.
| |
Collapse
|
262
|
Inflammation-regulated mRNA stability and the progression of vascular inflammatory diseases. Clin Sci (Lond) 2017; 131:2687-2699. [PMID: 29109302 DOI: 10.1042/cs20171373] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 10/09/2017] [Accepted: 10/10/2017] [Indexed: 12/22/2022]
Abstract
Cardiovascular disease remains a major medical and socioeconomic burden in developed and developing societies, and will increase with an aging and increasingly sedentary society. Vascular disease and atherosclerotic vascular syndromes are essentially inflammatory disorders, and transcriptional and post-transcriptional processes play essential roles in the ability of resident vascular and inflammatory cells to adapt to environmental stimuli. The regulation of mRNA translocation, stability, and translation are key processes of post-transcriptional regulation that permit these cells to rapidly respond to inflammatory stimuli. For the most part, these processes are controlled by elements in the 3'-UTR of labile, proinflammatory transcripts. Since proinflammatory transcripts almost exclusively contain AU-rich elements (AREs), this represents a tightly regulated and specific mechanism for initiation and maintenance of the proinflammatory phenotype. RNA-binding proteins (RBPs) recognize cis elements in 3'-UTR, and regulate each of these processes, but there is little literature exploring the concept that RBPs themselves can be directly regulated by inflammatory stimuli. Conceptually, inflammation-responsive RBPs represent an attractive target of rational therapies to combat vascular inflammatory syndromes. Herein we briefly describe the cellular and molecular etiology of atherosclerosis, and summarize our current understanding of RBPs and their specific roles in regulation of inflammatory mRNA stability. We also detail RBPs as targets of current anti-inflammatory modalities and how this may translate into better treatment for vascular inflammatory diseases.
Collapse
|
263
|
van Poelgeest EP, Dillingh MR, de Kam M, Malone KE, Kemper M, Stroes ESG, Burggraaf J, Moerland M. Characterization of immune cell, endothelial, and renal responses upon experimental human endotoxemia. J Pharmacol Toxicol Methods 2017; 89:39-46. [PMID: 29056520 DOI: 10.1016/j.vascn.2017.10.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 10/06/2017] [Accepted: 10/11/2017] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Although the effects of relatively high concentrations of endotoxin on endothelial activation/dysfunction and kidney markers has been described in literature, detailed insight in the LPS concentration-effect relationship, the magnitude, variability and timing of the response, and potential effects of endotoxemia on the kidneys is lacking. A study was performed to assess the effects of low- to moderate dose (0.5, 1 or 2ng/kg) endotoxemia on the endothelium and kidneys as measured by a panel of novel highly sensitive kidney injury markers. METHODS This was a randomized, double-blind, placebo-controlled study with single ascending doses of LPS (0.5, 1 or 2ng/kg) administered to healthy male volunteers (3 cohorts of 8 subjects, LPS:placebo 6:2). Endothelial measures included selectins, cell adhesion molecules, and thrombomodulin. Renal measures included novel, sensitive and specific biomarkers of acute kidney injury. RESULTS Endotoxin exposure resulted in consistent LPS dose-dependent responses in inflammatory markers, E- and P- Selectin, VCAM1, ICAM1, and thrombomodulin. The observed biological responses were transient, reaching a level of significance of at least <0.01 in the highest dose group and with an effect size which was dependent on the administered LPS dose. LPS-induced inflammatory and endothelial effects did not translate into a change in renal damage biomarkers, although at 2ng/kg LPS, subtle and transient biomarker changes were observed that may relate to (subclinical) tubular damage. DISCUSSION We demonstrated that administration of a single LPS dose of 2ng/kg to healthy volunteers results in significant inflammatory and endothelial responses, without inducing clinically relevant signs of kidney injury. These findings support the application of the human endotoxemia model in future clinical pharmacology studies.
Collapse
Affiliation(s)
| | - Marlous R Dillingh
- Centre for Human Drug Research, Zernikedreef 8, 2333, CL, Leiden, The Netherlands.
| | - Marieke de Kam
- Centre for Human Drug Research, Zernikedreef 8, 2333, CL, Leiden, The Netherlands.
| | - Karen E Malone
- Good Biomarker Sciences, Zernikedreef 8, 2333, CL, Leiden, The Netherlands.
| | - Marleen Kemper
- Academic Medical Center, Meibergdreef 9, 1105, AZ, Amsterdam, The Netherlands.
| | - Erik S G Stroes
- Academic Medical Center, Meibergdreef 9, 1105, AZ, Amsterdam, The Netherlands.
| | - Jacobus Burggraaf
- Centre for Human Drug Research, Zernikedreef 8, 2333, CL, Leiden, The Netherlands.
| | - Matthijs Moerland
- Centre for Human Drug Research, Zernikedreef 8, 2333, CL, Leiden, The Netherlands.
| |
Collapse
|
264
|
Jang SA, Park DW, Kwon JE, Song HS, Park B, Jeon H, Sohn EH, Koo HJ, Kang SC. Quinic acid inhibits vascular inflammation in TNF-α-stimulated vascular smooth muscle cells. Biomed Pharmacother 2017; 96:563-571. [PMID: 29032340 DOI: 10.1016/j.biopha.2017.10.021] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 09/11/2017] [Accepted: 10/02/2017] [Indexed: 02/04/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease, and the increased expression of adhesion molecules on vascular smooth muscle cells contributes to the progression of vascular disease. Quinic acid (QA) has been shown to possess radioprotection, anti-neuroinflammatory, and anti-oxidant activities; however, an anti-vascular inflammatory effect has not been reported. This study investigated the effect of QA on the expression of vascular cell adhesion molecule-1 (VCAM-1) stimulated by TNF-α in MOVAS cells. Pre-incubation of MOVAS cells, the mouse vascular smooth muscle cell line for 2h with QA (0.1, 1 and 10 μg/mL) dose-dependently inhibits TNF-α-induced mRNA and protein expression of VCAM-1 and monocyte adhesion. QA inhibits TNF-α-stimulated phosphorylation of MAP kinase and NK-κB activation. Our results indicate that QA inhibits the TNF-α-stimulated induction of VCAM-1 in VSMC by inhibiting the MAP kinase and NF-κB signaling pathways and the adhesion capacity of VSMC, which may explain the ability of QA to inhibit vascular inflammation such as atherosclerosis.
Collapse
Affiliation(s)
- Seon-A Jang
- Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Dae Won Park
- Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Jeong Eun Kwon
- Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Hae Seong Song
- Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Bongkyun Park
- Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Hyelin Jeon
- Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Eun-Hwa Sohn
- Department of Herbal Medicine Resources, Kangwon National University, Samcheok 25913, Republic of Korea
| | - Hyun Jung Koo
- Department of Medicinal and Industrial Crops, Korea National College of Agriculture and Fisheries, Jeonju 54874, Republic of Korea
| | - Se Chan Kang
- Department of Oriental Medicine Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, Republic of Korea.
| |
Collapse
|
265
|
Cytokine-induced autophagy promotes long-term VCAM-1 but not ICAM-1 expression by degrading late-phase IκBα. Sci Rep 2017; 7:12472. [PMID: 28963466 PMCID: PMC5622139 DOI: 10.1038/s41598-017-12641-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 09/13/2017] [Indexed: 12/31/2022] Open
Abstract
Pro-inflammatory cytokines are known to induce endothelial cell autophagy, but the role of autophagy in regulating the expression of pro-inflammatory molecules has not been characterized. We hypothesized that autophagy facilitates expression of endothelial adhesion molecules. TNFα and IL-1β induced autophagy markers in human umbilical vein endothelial cells and inhibition of autophagy by 3-methyladenine (3-MA) blocked adhesion of Jurkat lymphocytes. Interestingly, 3-MA suppressed VCAM-1 but not ICAM-1 expression at 24 hours but not 6 hours. 3-MA suppressed VCAM-1 transcription and decreased nuclear NF-κB p65 level at 6 hours but not at 2 hours. Cytokines induced a biphasic degradation of IκBα and 3-MA selectively blocked the late-phase IκBα degradation. Our results suggest that cytokine-induced autophagy contributes to late-phase IκBα degradation, facilitates NF-κB nuclear translocation and VCAM-1 transcription for long-term VCAM-1 expression. With a cytokines array assay, we found that 3-MA also inhibited IP-10 expression. These findings provide new information about the role of endothelial autophagy in persistent expression of VCAM-1 and IP-10 which enhance lymphocyte recruitment and adhesion to endothelium.
Collapse
|
266
|
Shon SM, Jang HJ, Schellingerhout D, Kim JY, Ryu WS, Lee SK, Kim J, Park JY, Oh JH, Kang JW, Je KH, Park JE, Kim K, Kwon IC, Lee J, Nahrendorf M, Park JH, Kim DE. Cytokine Response to Diet and Exercise Affects Atheromatous Matrix Metalloproteinase-2/9 Activity in Mice. Circ J 2017; 81:1528-1536. [PMID: 28883215 DOI: 10.1253/circj.cj-16-1196] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2024]
Abstract
BACKGROUND The aim of this study is to identify the principal circulating factors that modulate atheromatous matrix metalloproteinase (MMP) activity in response to diet and exercise. METHODS AND RESULTS Apolipoprotein-E knock-out (ApoE-/-) mice (n=56) with pre-existing plaque, fed either a Western diet (WD) or normal diet (ND), underwent either 10 weeks of treadmill exercise or had no treatment. Atheromatous MMP activity was visualized using molecular imaging with a MMP-2/9 activatable near-infrared fluorescent (NIRF) probe. Exercise did not significantly reduce body weight, visceral fat, and plaque size in either WD-fed animals or ND-fed animals. However, atheromatous MMP-activity was different; ND animals that did or did not exercise had similarly low MMP activities, WD animals that did not exercise had high MMP activity, and WD animals that did exercise had reduced levels of MMP activity, close to the levels of ND animals. Factor analysis and path analysis showed that soluble vascular cell adhesion molecule (sVCAM)-1 was directly positively correlated to atheromatous MMP activity. Adiponectin was indirectly negatively related to atheromatous MMP activity by way of sVCAM-1. Resistin was indirectly positively related to atheromatous MMP activity by way of sVCAM-1. Visceral fat amount was indirectly positively associated with atheromatous MMP activity, by way of adiponectin reduction and resistin elevation. MMP-2/9 imaging of additional mice (n=18) supported the diet/exercise-related anti-atherosclerotic roles for sVCAM-1. CONCLUSIONS Diet and exercise affect atheromatous MMP activity by modulating the systemic inflammatory milieu, with sVCAM-1, resistin, and adiponectin closely interacting with each other and with visceral fat.
Collapse
Affiliation(s)
- Soo-Min Shon
- Molecular Imaging and Neurovascular Research (MINER) Laboratory, Dongguk University Ilsan Hospital
| | - Hee Jeong Jang
- Molecular Imaging and Neurovascular Research (MINER) Laboratory, Dongguk University Ilsan Hospital
- Department of Medical Biotechnology, Dongguk University
| | - Dawid Schellingerhout
- Departments of Diagnostic Radiology and Cancer Systems, University of Texas M.D. Anderson Cancer Center
| | - Jeong-Yeon Kim
- Molecular Imaging and Neurovascular Research (MINER) Laboratory, Dongguk University Ilsan Hospital
| | - Wi-Sun Ryu
- Molecular Imaging and Neurovascular Research (MINER) Laboratory, Dongguk University Ilsan Hospital
| | - Su-Kyoung Lee
- Molecular Imaging and Neurovascular Research (MINER) Laboratory, Dongguk University Ilsan Hospital
| | - Jiwon Kim
- Molecular Imaging and Neurovascular Research (MINER) Laboratory, Dongguk University Ilsan Hospital
| | - Jin-Yong Park
- Molecular Imaging and Neurovascular Research (MINER) Laboratory, Dongguk University Ilsan Hospital
| | - Ji Hye Oh
- Molecular Imaging and Neurovascular Research (MINER) Laboratory, Dongguk University Ilsan Hospital
| | - Jeong Wook Kang
- Molecular Imaging and Neurovascular Research (MINER) Laboratory, Dongguk University Ilsan Hospital
| | - Kang-Hoon Je
- Molecular Imaging and Neurovascular Research (MINER) Laboratory, Dongguk University Ilsan Hospital
| | - Jung E Park
- Molecular Imaging and Neurovascular Research (MINER) Laboratory, Dongguk University Ilsan Hospital
| | - Kwangmeyung Kim
- Biomedical Research Center, Korea Institute of Science and Technology
| | - Ick Chan Kwon
- Biomedical Research Center, Korea Institute of Science and Technology
| | - Juneyoung Lee
- Department of Biostatistics, College of Medicine, Korea University
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School
| | | | - Dong-Eog Kim
- Molecular Imaging and Neurovascular Research (MINER) Laboratory, Dongguk University Ilsan Hospital
| |
Collapse
|
267
|
Nicolaou A, Northoff BH, Sass K, Ernst J, Kohlmaier A, Krohn K, Wolfrum C, Teupser D, Holdt LM. Quantitative trait locus mapping in mice identifies phospholipase Pla2g12a as novel atherosclerosis modifier. Atherosclerosis 2017; 265:197-206. [PMID: 28917158 DOI: 10.1016/j.atherosclerosis.2017.08.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 08/23/2017] [Accepted: 08/24/2017] [Indexed: 01/06/2023]
Abstract
BACKGROUND AND AIMS In a previous work, a female-specific atherosclerosis risk locus on chromosome (Chr) 3 was identified in an intercross of atherosclerosis-resistant FVB and atherosclerosis-susceptible C57BL/6 (B6) mice on the LDL-receptor deficient (Ldlr-/-) background. It was the aim of the current study to identify causative genes at this locus. METHODS We established a congenic mouse model, where FVB.Chr3B6/B6 mice carried an 80 Mb interval of distal Chr3 on an otherwise FVB.Ldlr-/- background, to validate the Chr3 locus. Candidate genes were identified using genome-wide expression analyses. Differentially expressed genes were validated using quantitative PCRs in F0 and F2 mice and their functions were investigated in pathophysiologically relevant cells. RESULTS Fine-mapping of the Chr3 locus revealed two overlapping, yet independent subloci for female atherosclerosis susceptibility: when transmitted by grandfathers to granddaughters, the B6 risk allele increased atherosclerosis and downregulated the expression of the secreted phospholipase Pla2g12a (2.6 and 2.2 fold, respectively); when inherited by grandmothers, the B6 risk allele induced vascular cell adhesion molecule 1 (Vcam1). Down-regulation of Pla2g12a and up-regulation of Vcam1 were validated in female FVB.Chr3B6/B6 congenic mice, which developed 2.5 greater atherosclerotic lesions compared to littermate controls (p=0.039). Pla2g12a was highly expressed in aortic endothelial cells in vivo, and knocking-down Pla2g12a expression by RNAi in cultured vascular endothelial cells or macrophages increased their adhesion to ECs in vitro. CONCLUSIONS Our data establish Pla2g12a as an atheroprotective candidate gene in mice, where high expression levels in ECs and macrophages may limit the recruitment and accumulation of these cells in nascent atherosclerotic lesions.
Collapse
Affiliation(s)
- Alexandros Nicolaou
- Institute of Laboratory Medicine, Ludwig Maximilians University Munich, Munich, Germany
| | - Bernd H Northoff
- Institute of Laboratory Medicine, Ludwig Maximilians University Munich, Munich, Germany
| | - Kristina Sass
- Institute of Laboratory Medicine, Ludwig Maximilians University Munich, Munich, Germany
| | - Jana Ernst
- Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Alexander Kohlmaier
- Institute of Laboratory Medicine, Ludwig Maximilians University Munich, Munich, Germany
| | - Knut Krohn
- Interdisciplinary Center for Clinical Research Leipzig (IZKF), Core-Unit DNA Technologies, University of Leipzig, Leipzig, Germany
| | - Christian Wolfrum
- Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
| | - Daniel Teupser
- Institute of Laboratory Medicine, Ludwig Maximilians University Munich, Munich, Germany
| | - Lesca M Holdt
- Institute of Laboratory Medicine, Ludwig Maximilians University Munich, Munich, Germany.
| |
Collapse
|
268
|
Akbar N, Digby JE, Cahill TJ, Tavare AN, Corbin AL, Saluja S, Dawkins S, Edgar L, Rawlings N, Ziberna K, McNeill E, Oxford Acute Myocardial Infarction (OxAMI) Study, 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: 77] [Impact Index Per Article: 9.6] [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
|
269
|
Brief Report: Macrophage Activation in HIV-Infected Adolescent Males Contributes to Differential Bone Loss by Sex: Adolescent Trials Network Study 021. J Acquir Immune Defic Syndr 2017; 72:372-5. [PMID: 26885808 DOI: 10.1097/qai.0000000000000953] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Accumulating evidence suggests that rates of low bone mass are greater in HIV-infected males than females. Of 11 biomarkers assessed by sex and HIV-status, HIV-infected males had increased levels of soluble CD14 which inversely correlated with bone mineral content and bone mineral density measures, suggesting macrophage activation as a possible mechanism of differential bone loss.
Collapse
|
270
|
Rizwan H, Mohanta J, Si S, Pal A. Gold nanoparticles reduce high glucose-induced oxidative-nitrosative stress regulated inflammation and apoptosis via tuberin-mTOR/NF-κB pathways in macrophages. Int J Nanomedicine 2017; 12:5841-5862. [PMID: 28860752 PMCID: PMC5566318 DOI: 10.2147/ijn.s141839] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hyperglycemia is a risk factor for cardiovascular mortality and morbidity, and directly responsible for exacerbating macrophage activation and atherosclerosis. We showed that gold nanoparticles (AuNPs) reduce the high glucose (HG)-induced atherosclerosis-related complications in macrophages via oxidative-nitrosative stress-regulated inflammation and apoptosis. The effects of AuNPs on oxidative-nitrosative stress markers such as cellular antioxidants were attenuated by HG exposure, leading to reduction in the accumulation of reactive oxygen/nitrogen species in cellular compartments. Further, these abnormalities of antioxidants level and reactive oxygen/nitrogen species accumulations initiate cellular stress, resulting in the activation of nuclear factor κB (NF-κB) via ERK1/2mitogen-activated protein kinase (MAPK)/Akt/tuberin-mammalian target of rapamycin (mTOR) pathways. The activated NF-κB stimulates inflammatory mediators, which subsequently subdue biomolecules damage, leading to aggravation of the inflammatory infiltration and immune responses. Treatment of AuNPs inhibits the intracellular redox-sensitive signaling pathways, inflammation, and apoptosis in macrophages. Together, our results indicate that AuNPs may modulate HG-induced oxidative-nitrosative stress. These effects may be sealed tight due to the fact that AuNPs treatment reduces the activation of NF-κB by ERK1/2MAPK/Akt/tuberin-mTOR pathways-mediated inflammatory genes expression and cellular stress responses, which may be beneficial for minimizing the atherosclerosis.
Collapse
Affiliation(s)
- Huma Rizwan
- School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Jagdeep Mohanta
- School of Applied Sciences, KIIT University, Bhubaneswar, India
| | - Satyabrata Si
- School of Biotechnology, KIIT University, Bhubaneswar, India.,School of Applied Sciences, KIIT University, Bhubaneswar, India
| | - Arttatrana Pal
- Department of Zoology, School of Life Sciences, Mahatma Gandhi Central University, Bihar, India
| |
Collapse
|
271
|
Abstract
Cardiovascular disease (CVD) is a major health problem globally. The high incidence and case fatality of CVD are, to a large extent, a consequence of its late diagnosis and lack of highly sensitive and specific markers. Only a very small number of biomarkers, such as troponin, detect late disease. There is some evidence of an association and dysregulation between specific cytokines in the pathogenesis of CVD. These molecules are involved in inflammatory and immune mechanisms associated with atherogenesis. Several molecular/cellular pathways that include STAT, MAPK, and SMAD are modulated by cytokines. Against this background, microRNAs (miRNAs) are a class of noncoding RNAs with important roles in pathological events, leading to atherosclerotic CVD. It has been shown that the latter could affect cytokine production and contribute to progression of atherosclerotic CVD. Moreover, modulation of miRNAs appears to inhibit cardiomyocyte apoptosis, attenuate infarct size, and reduce cardiac dysfunction. This review highlights several recent preclinical and clinical studies on the role of cytokines in CVD, novel miRNA-based therapeutic approaches for therapeutic intervention, and potential circulating cytokines that have promise as biomarkers in CVD.
Collapse
Affiliation(s)
- Hamed Mirzaei
- School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Sussex, United Kingdom
| | - Amir Avan
- School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Majid G Mobarhan
- School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
272
|
Chang AH, Raftrey BC, D'Amato G, Surya VN, Poduri A, Chen HI, Goldstone AB, Woo J, Fuller GG, Dunn AR, Red-Horse K. DACH1 stimulates shear stress-guided endothelial cell migration and coronary artery growth through the CXCL12-CXCR4 signaling axis. Genes Dev 2017; 31:1308-1324. [PMID: 28779009 PMCID: PMC5580653 DOI: 10.1101/gad.301549.117] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 06/30/2017] [Indexed: 01/03/2023]
Abstract
Sufficient blood flow to tissues relies on arterial blood vessels, but the mechanisms regulating their development are poorly understood. Many arteries, including coronary arteries of the heart, form through remodeling of an immature vascular plexus in a process triggered and shaped by blood flow. However, little is known about how cues from fluid shear stress are translated into responses that pattern artery development. Here, we show that mice lacking endothelial Dach1 had small coronary arteries, decreased endothelial cell polarization, and reduced expression of the chemokine Cxcl12 Under shear stress in culture, Dach1 overexpression stimulated endothelial cell polarization and migration against flow, which was reversed upon CXCL12/CXCR4 inhibition. In vivo, DACH1 was expressed during early arteriogenesis but was down in mature arteries. Mature artery-type shear stress (high, uniform laminar) specifically down-regulated DACH1, while the remodeling artery-type flow (low, variable) maintained DACH1 expression. Together, our data support a model in which DACH1 stimulates coronary artery growth by activating Cxcl12 expression and endothelial cell migration against blood flow into developing arteries. This activity is suppressed once arteries reach a mature morphology and acquire high, laminar flow that down-regulates DACH1. Thus, we identified a mechanism by which blood flow quality balances artery growth and maturation.
Collapse
Affiliation(s)
- Andrew H Chang
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California 94305, USA
- Department of Biology, Stanford University, Stanford, California 94305, USA
| | - Brian C Raftrey
- Department of Biology, Stanford University, Stanford, California 94305, USA
| | - Gaetano D'Amato
- Department of Biology, Stanford University, Stanford, California 94305, USA
| | - Vinay N Surya
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, USA
| | - Aruna Poduri
- Department of Biology, Stanford University, Stanford, California 94305, USA
| | - Heidi I Chen
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California 94305, USA
- Department of Biology, Stanford University, Stanford, California 94305, USA
| | - Andrew B Goldstone
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, California 94305, USA
- Department of Health Research and Policy-Epidemiology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Gerald G Fuller
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, USA
| | - Alexander R Dunn
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, USA
| | - Kristy Red-Horse
- Department of Biology, Stanford University, Stanford, California 94305, USA
| |
Collapse
|
273
|
Boese AC, Kim SC, Yin KJ, Lee JP, Hamblin MH. Sex differences in vascular physiology and pathophysiology: estrogen and androgen signaling in health and disease. Am J Physiol Heart Circ Physiol 2017. [PMID: 28626075 DOI: 10.1152/ajpheart.00217.2016] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Sex differences between women and men are often overlooked and underappreciated when studying the cardiovascular system. It has been long assumed that men and women are physiologically similar, and this notion has resulted in women being clinically evaluated and treated for cardiovascular pathophysiological complications as men. Currently, there is increased recognition of fundamental sex differences in cardiovascular function, anatomy, cell signaling, and pathophysiology. The National Institutes of Health have enacted guidelines expressly to gain knowledge about ways the sexes differ in both normal function and diseases at the various research levels (molecular, cellular, tissue, and organ system). Greater understanding of these sex differences will be used to steer future directions in the biomedical sciences and translational and clinical research. This review describes sex-based differences in the physiology and pathophysiology of the vasculature, with a special emphasis on sex steroid receptor (estrogen and androgen receptor) signaling and their potential impact on vascular function in health and diseases (e.g., atherosclerosis, hypertension, peripheral artery disease, abdominal aortic aneurysms, cerebral aneurysms, and stroke).
Collapse
Affiliation(s)
- Austin C Boese
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Seong C Kim
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Ke-Jie Yin
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jean-Pyo Lee
- Department of Neurology, Tulane University School of Medicine, New Orleans, Louisiana; and.,Center for Stem Cell Research and Regenerative Medicine, New Orleans, Louisiana
| | - Milton H Hamblin
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana;
| |
Collapse
|
274
|
Li S, Ning H, Ye Y, Wei W, Guo R, Song Q, Liu L, Liu Y, Na L, Niu Y, Chu X, Feng R, Moustaid-Moussa N, Li Y, Sun C. Increasing extracellular Ca 2+ sensitizes TNF-alpha-induced vascular cell adhesion molecule-1 (VCAM-1) via a TRPC1/ERK1/2/NFκB-dependent pathway in human vascular endothelial cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1566-1577. [PMID: 28583863 DOI: 10.1016/j.bbamcr.2017.06.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 05/16/2017] [Accepted: 06/01/2017] [Indexed: 12/29/2022]
Abstract
Increasing circulating Ca2+ levels within the normal range has been reported to positively correlate with the incidence of fatal cardiovascular diseases (CVDs). However, limited studies have been able to delineate the potential mechanism(s) linking circulating Ca2+ to CVD. In this study, we exposed primary human umbilical vein endothelial cells (HUVECs) and human umbilical vein cell line (EA.hy926) to different extracellular Ca2+ to mimic the physiological state. Our data revealed that increasing extracellular Ca2+ significantly enhanced susceptibility to tumor necrosis factor (TNF)-alpha-stimulated vascular cell adhesion molecule (VCAM)-1 expression and monocytes adhesion. Knocking-down VCAM-1 by siRNA abolished calcium-induced monocytes adhesion on HUVECs. Follow up mechanistic investigations identified that extracellular Ca2+-increased calcium influx contributed to the activation of VCAM-1. This was mediated via upregulation of transient receptor potential channel (TRPC)1 in a nuclear factor (NF)κB-dependent manner. Most importantly, we found that a novel TRPC1-regulated extracellular signal-regulated kinase 1/2 (ERK1/2) pathway exclusively contributed to calcium-induced NFκB activation. This study provided direct evidence that increasing extracellular Ca2+ enhanced TNF-alpha-induced VCAM-1 activation and monocytes adhesion. Moreover, we identified a novel TRPC1/ERK1/2/NFκB signaling pathway mediating VCAM-1 activation and monocyte adhesion in this pathological process. Our studies indicate that blood calcium levels should be strictly monitored to help prevent CVD, and that TRPC1 might act as a potential target for the treatment and prevention against increased circulating calcium-enhanced CVDs.
Collapse
Affiliation(s)
- Songtao Li
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin 150081, China; Research Institute of Food, Nutrition and Health, Sino-Russian Medical Research Center, Harbin Medical University, Harbin 150081, China; Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University, Ministry of Education, 150081, China
| | - Hua Ning
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin 150081, China; Research Institute of Food, Nutrition and Health, Sino-Russian Medical Research Center, Harbin Medical University, Harbin 150081, China
| | - Yaxin Ye
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Wei Wei
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Rui Guo
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Qing Song
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Lei Liu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Yunyun Liu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Lixin Na
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin 150081, China; Research Institute of Food, Nutrition and Health, Sino-Russian Medical Research Center, Harbin Medical University, Harbin 150081, China
| | - Yuchun Niu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin 150081, China; Research Institute of Food, Nutrition and Health, Sino-Russian Medical Research Center, Harbin Medical University, Harbin 150081, China
| | - Xia Chu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin 150081, China; Research Institute of Food, Nutrition and Health, Sino-Russian Medical Research Center, Harbin Medical University, Harbin 150081, China
| | - Rennan Feng
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin 150081, China; Research Institute of Food, Nutrition and Health, Sino-Russian Medical Research Center, Harbin Medical University, Harbin 150081, China
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences and Obesity Research Cluster, Texas Tech University, Lubbock, TX 79409, USA
| | - Ying Li
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin 150081, China; Research Institute of Food, Nutrition and Health, Sino-Russian Medical Research Center, Harbin Medical University, Harbin 150081, China.
| | - Changhao Sun
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin 150081, China; Research Institute of Food, Nutrition and Health, Sino-Russian Medical Research Center, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
275
|
Lu Y, Zhang M, Zhao P, Jia M, Liu B, Jia Q, Guo J, Dou L, Li J. Modified citrus pectin inhibits galectin-3 function to reduce atherosclerotic lesions in apoE-deficient mice. Mol Med Rep 2017; 16:647-653. [PMID: 28560429 PMCID: PMC5482107 DOI: 10.3892/mmr.2017.6646] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 01/27/2017] [Indexed: 12/18/2022] Open
Abstract
Galectin-3 is a carbohydrate-binding lectin, which has been implicated in the modulation of atherosclerotic pathophysiology, and is highly expressed in monocytes, macrophages and endothelial cells within atherosclerotic plaques. Modified citrus pectin (MCP) is produced from citrus pectin via pH and temperature modifications, which break it into shorter, non‑branched, galactose‑rich carbohydrate chains. MCP is able to tightly bind with galectin‑3, via recognition of its carbohydrate recognition domain, and facilitates the modulation of galectin‑3‑induced bioactivity. The present study explored the effects of MCP on the initiation of atherosclerosis. Eight‑week‑old apolipoprotein E‑deficient mice were treated with 1% MCP and fed an atherogenic diet for 4 weeks. The effects of MCP on atherosclerotic initiation were determined by pathological analysis and scanning electron microscope (SEM) imaging. MCP treatment reduced the size of atherosclerotic lesion areas, which was accompanied by decreased numbers of macrophages and smooth muscle cells (SMCs). Furthermore, SEM examination of the surface of the atheroma‑prone vessel wall indicated that MCP treatment reduced endothelial injury. To analyze the effects of MCP on monocyte adhesion, firstly, oxidized‑low density lipoprotein and various concentrations of MCP (0.025, 0.05, 0.1 and 0.25%) were incubated with the human umbilical vein endothelial cells (HUVECs) for stimulation and following this, the U937 cells were plated onto the HUVECs. The results revealed that MCP reduced the adhesion of U937 monocytes to HUVECs, indicating the adhesion-inhibiting effects of MCP. In conclusion, the present study revealed that MCP, a galectin‑3 inhibitor, reduced the size of atherosclerotic lesions by inhibiting the adhesion of leucocytes to endothelial cells. Inhibition of galectin‑3 function may be a therapeutic strategy for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Yonggang Lu
- Clinical Laboratory, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| | - Mingming Zhang
- Clinical Laboratory, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| | - Pei Zhao
- Clinical Laboratory, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| | - Min Jia
- Clinical Laboratory, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| | - Bing Liu
- Neurology Department 2, Handan Central Hospital, Handan, Hebei 056001, P.R. China
| | - Qian Jia
- Clinical Laboratory, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| | - Jun Guo
- Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, Ministry of Health, Beijing 100730, P.R. China
| | - Lin Dou
- Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, Ministry of Health, Beijing 100730, P.R. China
| | - Jian Li
- Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, Ministry of Health, Beijing 100730, P.R. China
| |
Collapse
|
276
|
Ungprasert P, Crowson CS, Matteson EL. Epidemiology and clinical characteristics of sarcoidosis: an update from a population-based cohort study from Olmsted County, Minnesota. Reumatismo 2017; 69:16-22. [PMID: 28535617 PMCID: PMC5521258 DOI: 10.4081/reumatismo.2017.965] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/27/2017] [Accepted: 03/30/2017] [Indexed: 12/22/2022] Open
Abstract
Information about the epidemiology, clinical manifestations and comorbidities of sarcoidosis among Caucasians is relatively scarce. This review focuses primarily on the data from a recently published Caucasianpredominant population-based cohort from Olmsted County, Minnesota. Overall, the incidence rate was 10.0 per 100,000 population, which suggested that sarcoidosis is less common in Caucasians than in Blacks, but is more common in Caucasians than in Asians. Intrathoracic involvement was seen in the vast majority of patients, but less than half have respiratory symptoms. The most common extra-thoracic manifestations were skin rash followed by arthralgia, ophthalmologic involvement, hepatic involvement, splenomegaly, renal involvement, neurological involvement, extra-thoracic lymphadenopathy, exocrine gland involvement, upper respiratory tract involvement and cardiac involvement. Compared to sex and age-matched subjects, patients with sarcoidosis suffer from increased rates of cardiovascular disease, venous thromboembolism and hospitalized infection.
Collapse
Affiliation(s)
- P Ungprasert
- Division of Rheumatology, Department of Internal Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN.
| | | | | |
Collapse
|
277
|
Finney AC, Funk SD, Green JM, Yurdagul A, Rana MA, Pistorius R, Henry M, Yurochko A, Pattillo CB, Traylor JG, Chen J, Woolard MD, Kevil CG, Orr AW. EphA2 Expression Regulates Inflammation and Fibroproliferative Remodeling in Atherosclerosis. Circulation 2017; 136:566-582. [PMID: 28487392 DOI: 10.1161/circulationaha.116.026644] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 05/03/2017] [Indexed: 01/24/2023]
Abstract
BACKGROUND Atherosclerotic plaque formation results from chronic inflammation and fibroproliferative remodeling in the vascular wall. We previously demonstrated that both human and mouse atherosclerotic plaques show elevated expression of EphA2, a guidance molecule involved in cell-cell interactions and tumorigenesis. METHODS Here, we assessed the role of EphA2 in atherosclerosis by deleting EphA2 in a mouse model of atherosclerosis (Apoe-/-) and by assessing EphA2 function in multiple vascular cell culture models. After 8 to 16 weeks on a Western diet, male and female mice were assessed for atherosclerotic burden in the large vessels, and plasma lipid levels were analyzed. RESULTS Despite enhanced weight gain and plasma lipid levels compared with Apoe-/- controls, EphA2-/-Apoe-/- knockout mice show diminished atherosclerotic plaque formation, characterized by reduced proinflammatory gene expression and plaque macrophage content. Although plaque macrophages express EphA2, EphA2 deletion does not affect macrophage phenotype, inflammatory responses, and lipid uptake, and bone marrow chimeras suggest that hematopoietic EphA2 deletion does not affect plaque formation. In contrast, endothelial EphA2 knockdown significantly reduces monocyte firm adhesion under flow. In addition, EphA2-/-Apoe-/- mice show reduced progression to advanced atherosclerotic plaques with diminished smooth muscle and collagen content. Consistent with this phenotype, EphA2 shows enhanced expression after smooth muscle transition to a synthetic phenotype, and EphA2 depletion reduces smooth muscle proliferation, mitogenic signaling, and extracellular matrix deposition both in atherosclerotic plaques and in vascular smooth muscle cells in culture. CONCLUSIONS Together, these data identify a novel role for EphA2 in atherosclerosis, regulating both plaque inflammation and progression to advanced atherosclerotic lesions. Cell culture studies suggest that endothelial EphA2 contributes to atherosclerotic inflammation by promoting monocyte firm adhesion, whereas smooth muscle EphA2 expression may regulate the progression to advanced atherosclerosis by regulating smooth muscle proliferation and extracellular matrix deposition.
Collapse
Affiliation(s)
- Alexandra C Finney
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.)
| | - Steven D Funk
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.)
| | - Jonette M Green
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.)
| | - Arif Yurdagul
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.)
| | - Mohammad Atif Rana
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.)
| | - Rebecca Pistorius
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.)
| | - Miriam Henry
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.)
| | - Andrew Yurochko
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.)
| | - Christopher B Pattillo
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.)
| | - James G Traylor
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.)
| | - Jin Chen
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.)
| | - Matthew D Woolard
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.)
| | - Christopher G Kevil
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.)
| | - A Wayne Orr
- From Departments of Cell Biology and Anatomy (A.C.F., S.D.F., J.M.G., A. Yurdagul, C.G.K., A.W.O.), Pathology and Translational Pathobiology (J.M.G., A. Yurdagul, R.P., M.H., J.G.T., C.G.K., A.W.O.), Cardiology (M.A.R.), Microbiology and Immunology (A. Yurochko, M.D.W.), and Molecular and Cellular Physiology (C.B.P., C.G.K., A.W.O.), Louisiana State University Health Sciences Center-Shreveport; Departments of Cancer Biology and Cell and Developmental Biology, Vanderbilt University, Nashville, TN (J.C.); and Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville (J.C.).
| |
Collapse
|
278
|
Zhang R. Ghrelin suppresses inflammation in HUVECs by inhibiting ubiquitin-mediated uncoupling protein 2 degradation. Int J Mol Med 2017; 39:1421-1427. [PMID: 28487946 PMCID: PMC5428956 DOI: 10.3892/ijmm.2017.2977] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 01/17/2017] [Indexed: 12/31/2022] Open
Abstract
Atherosclerosis is considered the major cause of heart attack, stroke and gangrene of the extremities, which is responsible for 50% of all mortality in Western countries. The pathogenesis and causes of atherosclerosis remain elusive. Recent studies highlight inflammation as a contributing factor for atherosclerosis in all stages of the disease process. In this study, we demonstrate that the treatment of human umbilical vein endothelial cells (HUVECs) with ghrelin inhibits the oxidized low-density lipoprotein (oxLDL)-induced inflammatory response, In addition, treatment with ghrelin led to the accumulation of uncoupling protein 2 (UCP2) in the cells, thus decreasing reactive oxygen species (ROS) generation. Moreover, the siRNA-mediated knockdown of UCP2 expression suggested that the inhibitory effects of ghrelin on the inflammatory response relied on its ability to induce the accumulation of cellular UCP2 levels. Further analysis indicated that the accumulation of UCP2 in the ghrelin-treated cells was due to the ability of ghrelin to inhibit the ubiquitination of UCP2 and prevent UCP2 degradation, resulting in the extended protein half-life of UCP2. On the whole, our data indicate that ghrelin inhibits the oxLDL-induced inflammatory response in HUVECs, and may thus have potential for use as an anti-atherosclerotic agent. Our data may also provide valuable insight into the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Ruolan Zhang
- Department of Cardiology, Harrison International Peace Hospital, Hengshui, Hebei 053000, P.R. China
| |
Collapse
|
279
|
Gao L, Fu S, Wang W, Xie C, Zhang Y, Gong F. Notch4 signaling pathway in a Kawasaki disease mouse model induced by Lactobacillus casei cell wall extract. Exp Ther Med 2017; 13:3438-3442. [PMID: 28587423 PMCID: PMC5450570 DOI: 10.3892/etm.2017.4434] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 02/17/2017] [Indexed: 01/01/2023] Open
Abstract
The present study aimed to explore the role of the Notch4 signaling pathway in a mouse model of Kawasaki disease (KD) induced by Lactobacillus casei cell wall extract (LCWE). BALB/c male mice (4–6 weeks old) were intraperitoneally injected with 500 µg LCWE in phosphate-buffered saline (PBS) or PBS alone (control group). At days 3, 7, 14 and 28, the numbers of circulating endothelial progenitor cells (EPCs) in the peripheral blood and the expression of Notch4 on the surface of EPCs were detected. In addition, the levels of vascular cell adhesion molecule 1 (VCAM-1) and P-selectin in the roots of coronary arteries were evaluated. The results demonstrated that the level of circulating EPCs increased significantly at day 3, decreased progressively from day 3 onwards, and recovered to the normal level at day 28. Furthermore, the expression of Notch4 on the surface of EPCs was evidently higher in the KD model compared with that in the control group at day 7. In the endothelial cells of the coronary artery root, the protein levels of VCAM-1 and P-selectin protein increased in the KD model. In conclusion, the Notch4 signaling pathway participated in the coronary artery lesions in the KD animal model induced by LCWE.
Collapse
Affiliation(s)
- Lichao Gao
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| | - Songling Fu
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| | - Wei Wang
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| | - Chunhong Xie
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| | - Yiying Zhang
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| | - Fangqi Gong
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
280
|
The inhibitory effects of polysaccharide peptides (PsP) of Ganoderma lucidum against atherosclerosis in rats with dyslipidemia. Heart Int 2017; 12:e1-e7. [PMID: 29114382 PMCID: PMC5460693 DOI: 10.5301/heartint.5000234] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2017] [Indexed: 01/01/2023] Open
Abstract
Background Atherosclerosis occurs as a result of low-density lipoprotein (LDL) deposits oxidation. Endothelial dysfunction is an early process of atherosclerosis. Restoring endothelial lining back to normal by endothelial progenitor cells (EPCs) is critical for slowing or reversing vascular disease progression. Oxidative stress from hydrogen peroxide (H2O2) is increased in dyslipidemia so that antioxidant agent is required to prevent destruction of blood vessels. Objectives This study aims to report Ganoderma lucidum polysaccharide peptide (PsP) effects in atherogenic process by measuring H2O2 level, IL-10 level, and EPC number in blood serum, and also intima-media thickness of aorta in dyslipidemia Wistar rat model by giving them a hypercholesterol diet (HCD). Materials and methods The study was an experimental in vivo post-test with control group design. Thirty-five Wistar rats (Rattus norwegicus) were divided into five groups (normal diet group, HCD group, and hypercholesterol groups that received 50 mg/kg, 150 mg/kg, and 300 mg/kg bodyweight PsP). Results Each treatment group showed significant results for the administration of PsP using the one-way analysis of variance test (p<0.050) for the reduction of H2O2 (p = 0.003), levels of IL-10 (p = 0.027), number of EPC in the blood serum (p = 0.011), and the intima-media thickness of the aorta (p = 0.000). PsP from G. lucidum is a potent antioxidant and may prevent atherogenesis process in patients with dyslipidemia. Conclusions The optimum doses of PsP in this study is 300 mg/kg bodyweight. Further studies are required to determine the antioxidant effects of PsP G. lucidum and its benefits in the management of dyslipidemia.
Collapse
|
281
|
Vogel ME, Idelman G, Konaniah ES, Zucker SD. Bilirubin Prevents Atherosclerotic Lesion Formation in Low-Density Lipoprotein Receptor-Deficient Mice by Inhibiting Endothelial VCAM-1 and ICAM-1 Signaling. J Am Heart Assoc 2017; 6:JAHA.116.004820. [PMID: 28365565 PMCID: PMC5532999 DOI: 10.1161/jaha.116.004820] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Numerous epidemiological studies support an inverse association between serum bilirubin levels and the incidence of cardiovascular disease; however, the mechanism(s) by which bilirubin may protect against atherosclerosis is undefined. The goals of the present investigations were to assess the ability of bilirubin to prevent atherosclerotic plaque formation in low‐density lipoprotein receptor‐deficient (Ldlr−/−) mice and elucidate the molecular processes underlying this effect. Methods and Results Bilirubin, at physiological concentrations (≤20 μmol/L), dose‐dependently inhibits THP‐1 monocyte migration across tumor necrosis factor α–activated human umbilical vein endothelial cell monolayers without altering leukocyte binding or cytokine production. A potent antioxidant, bilirubin effectively blocks the generation of cellular reactive oxygen species induced by the cross‐linking of endothelial vascular cell adhesion molecule 1 (VCAM‐1) or intercellular adhesion molecule 1 (ICAM‐1). These findings were validated by treating cells with blocking antibodies or with specific inhibitors of VCAM‐1 and ICAM‐1 signaling. When administered to Ldlr−/− mice on a Western diet, bilirubin (30 mg/kg intraperitoneally) prevents atherosclerotic plaque formation, but does not alter circulating cholesterol or chemokine levels. Aortic roots from bilirubin‐treated animals exhibit reduced lipid and collagen deposition, decreased infiltration of monocytes and lymphocytes, fewer smooth muscle cells, and diminished levels of chlorotyrosine and nitrotyrosine, without changes in VCAM‐1 or ICAM‐1 expression. Conclusions Bilirubin suppresses atherosclerotic plaque formation in Ldlr−/− mice by disrupting endothelial VCAM‐1‐ and ICAM‐1‐mediated leukocyte migration through the scavenging of reactive oxygen species signaling intermediaries. These findings suggest a potential mechanism for the apparent cardioprotective effects of bilirubin.
Collapse
Affiliation(s)
- Megan E Vogel
- Division of Digestive Diseases, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Gila Idelman
- Division of Digestive Diseases, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Eddy S Konaniah
- Department of Pathology and Laboratory Medicine, Metabolic Disease Institute, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Stephen D Zucker
- Division of Digestive Diseases, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
282
|
Eleftheriadis T, Pissas G, Sounidaki M, Antoniadi G, Rountas C, Liakopoulos V, Stefanidis L. Tryptophan depletion under conditions that imitate insulin resistance enhances fatty acid oxidation and induces endothelial dysfunction through reactive oxygen species-dependent and independent pathways. Mol Cell Biochem 2017; 428:41-56. [PMID: 28161804 DOI: 10.1007/s11010-016-2915-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 12/21/2016] [Indexed: 12/25/2022]
Abstract
In atherosclerosis-associated pathologic entities characterized by malnutrition and inflammation, L-tryptophan (TRP) levels are low. Insulin resistance is an independent cardiovascular risk factor and induces endothelial dysfunction by increasing fatty acid oxidation. It is also associated with inflammation and low TRP levels. Low TRP levels have been related to worse cardiovascular outcome. This study evaluated the effect of TRP depletion on endothelial dysfunction under conditions that imitate insulin resistance. Fatty acid oxidation, harmful pathways due to increased fatty acid oxidation, and endothelial dysfunction were assessed in primary human aortic endothelial cells cultured under normal glucose, low insulin conditions in the presence or absence of TRP. TRP depletion activated general control non-derepressible 2 kinase and inhibited aryl hydrocarbon receptor. It increased fatty acid oxidation by increasing expression and activity of carnitine palmitoyltransferase 1. Elevated fatty acid oxidation increased the formation of reactive oxygen species (ROS) triggering the polyol and hexosamine pathways, and enhancing protein kinase C activity and methylglyoxal production. TRP absence inhibited nitric oxide synthase activity in a ROS-dependent way, whereas it increased the expression of ICAM-1 and VCAM-1 in a ROS independent and possibly p53-dependent manner. Thus, TRP depletion, an amino acid whose low levels have been related to worse cardiovascular outcome and to inflammatory atherosclerosis-associated pathologic entities, under conditions that imitate insulin resistance enhances fatty acid oxidation and induces endothelial dysfunction through ROS-dependent and independent pathways. These findings may offer new insights at the molecular mechanisms involved in accelerated atherosclerosis that frequently accompanies malnutrition and inflammation.
Collapse
Affiliation(s)
- Theodoros Eleftheriadis
- Department of Nephrology, Medical School, University of Thessaly, Neo Ktirio, Mezourlo Hill, 41110, Larissa, Greece.
| | - Georgios Pissas
- Department of Nephrology, Medical School, University of Thessaly, Neo Ktirio, Mezourlo Hill, 41110, Larissa, Greece
| | - Maria Sounidaki
- Department of Nephrology, Medical School, University of Thessaly, Neo Ktirio, Mezourlo Hill, 41110, Larissa, Greece
| | - Georgia Antoniadi
- Department of Nephrology, Medical School, University of Thessaly, Neo Ktirio, Mezourlo Hill, 41110, Larissa, Greece
| | - Christos Rountas
- Department of Interventional Radiology, Medical School, University of Thessaly, Larissa, Greece
| | - Vassilios Liakopoulos
- Department of Nephrology, Medical School, University of Thessaly, Neo Ktirio, Mezourlo Hill, 41110, Larissa, Greece
| | - Loannis Stefanidis
- Department of Nephrology, Medical School, University of Thessaly, Neo Ktirio, Mezourlo Hill, 41110, Larissa, Greece
| |
Collapse
|
283
|
Leukocyte Trafficking in Cardiovascular Disease: Insights from Experimental Models. Mediators Inflamm 2017; 2017:9746169. [PMID: 28465628 PMCID: PMC5390637 DOI: 10.1155/2017/9746169] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 03/01/2017] [Indexed: 11/17/2022] Open
Abstract
Chemokine-induced leukocyte migration into the vessel wall is an early pathological event in the progression of atherosclerosis, the underlying cause of myocardial infarction. The immune-inflammatory response, mediated by both the innate and adaptive immune cells, is involved in the initiation, recruitment, and resolution phases of cardiovascular disease progression. Activation of leukocytes via inflammatory mediators such as chemokines, cytokines, and adhesion molecules is instrumental in these processes. In this review, we highlight leukocyte activation with the main focus being on the mechanisms of chemokine-mediated recruitment in atherosclerosis and the response postmyocardial infarction with key examples from experimental models of cardiovascular inflammation.
Collapse
|
284
|
Integrin signaling in atherosclerosis. Cell Mol Life Sci 2017; 74:2263-2282. [PMID: 28246700 DOI: 10.1007/s00018-017-2490-4] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/24/2017] [Accepted: 02/15/2017] [Indexed: 02/07/2023]
Abstract
Atherosclerosis, a chronic lipid-driven inflammatory disease affecting large arteries, represents the primary cause of cardiovascular disease in the world. The local remodeling of the vessel intima during atherosclerosis involves the modulation of vascular cell phenotype, alteration of cell migration and proliferation, and propagation of local extracellular matrix remodeling. All of these responses represent targets of the integrin family of cell adhesion receptors. As such, alterations in integrin signaling affect multiple aspects of atherosclerosis, from the earliest induction of inflammation to the development of advanced fibrotic plaques. Integrin signaling has been shown to regulate endothelial phenotype, facilitate leukocyte homing, affect leukocyte function, and drive smooth muscle fibroproliferative remodeling. In addition, integrin signaling in platelets contributes to the thrombotic complications that typically drive the clinical manifestation of cardiovascular disease. In this review, we examine the current literature on integrin regulation of atherosclerotic plaque development and the suitability of integrins as potential therapeutic targets to limit cardiovascular disease and its complications.
Collapse
|
285
|
Cortese R, Gileles-Hillel A, Khalyfa A, Almendros I, Akbarpour M, Khalyfa AA, Qiao Z, Garcia T, Andrade J, Gozal D. Aorta macrophage inflammatory and epigenetic changes in a murine model of obstructive sleep apnea: Potential role of CD36. Sci Rep 2017; 7:43648. [PMID: 28240319 PMCID: PMC5327416 DOI: 10.1038/srep43648] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 01/25/2017] [Indexed: 12/15/2022] Open
Abstract
Obstructive sleep apnea (OSA) affects 8-10% of the population, is characterized by chronic intermittent hypoxia (CIH), and causally associates with cardiovascular morbidities. In CIH-exposed mice, closely mimicking the chronicity of human OSA, increased accumulation and proliferation of pro-inflammatory metabolic M1-like macrophages highly expressing CD36, emerged in aorta. Transcriptomic and MeDIP-seq approaches identified activation of pro-atherogenic pathways involving a complex interplay of histone modifications in functionally-relevant biological pathways, such as inflammation and oxidative stress in aorta macrophages. Discontinuation of CIH did not elicit significant improvements in aorta wall macrophage phenotype. However, CIH-induced aorta changes were absent in CD36 knockout mice, Our results provide mechanistic insights showing that CIH exposures during sleep in absence of concurrent pro-atherogenic settings (i.e., genetic propensity or dietary manipulation) lead to the recruitment of CD36(+)high macrophages to the aortic wall and trigger atherogenesis. Furthermore, long-term CIH-induced changes may not be reversible with usual OSA treatment.
Collapse
Affiliation(s)
- Rene Cortese
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Alex Gileles-Hillel
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Abdelnaby Khalyfa
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Isaac Almendros
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Mahzad Akbarpour
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Ahamed A Khalyfa
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Zhuanghong Qiao
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Tzintzuni Garcia
- Center for Research Informatics, The University of Chicago, Chicago, IL, USA
| | - Jorge Andrade
- Center for Research Informatics, The University of Chicago, Chicago, IL, USA
| | - David Gozal
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
286
|
Ig-like domain 6 of VCAM-1 is a potential therapeutic target in TNFα-induced angiogenesis. Exp Mol Med 2017; 49:e294. [PMID: 28209985 PMCID: PMC5336558 DOI: 10.1038/emm.2016.147] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 10/07/2016] [Accepted: 10/14/2016] [Indexed: 01/05/2023] Open
Abstract
Tumor necrosis factor alpha (TNFα)-induced angiogenesis plays important roles in the progression of various diseases, including cancer, wet age-related macular degeneration, and rheumatoid arthritis. However, the relevance and role of vascular cell adhesion molecule-1 (VCAM-1) in angiogenesis have not yet been clearly elucidated. In this study, VCAM-1 knockdown shows VCAM-1 involvement in TNFα-induced angiogenesis. Through competitive blocking experiments with VCAM-1 Ig-like domain 6 (VCAM-1-D6) protein, we identified VCAM-1-D6 as a key domain regulating TNFα-induced vascular tube formation. We demonstrated that a monoclonal antibody specific to VCAM-1-D6 suppressed TNFα-induced endothelial cell migration and tube formation and TNFα-induced vessel sprouting in rat aortas. We also found that the antibody insignificantly affected endothelial cell viability, morphology and activation. Finally, the antibody specifically blocked VCAM-1-mediated cell–cell contacts by directly inhibiting VCAM-1-D6-mediated interaction between VCAM-1 molecules. These findings suggest that VCAM-1-D6 may be a potential novel therapeutic target in TNFα-induced angiogenesis and that antibody-based modulation of VCAM-1-D6 may be an effective strategy to suppress TNFα-induced angiogenesis.
Collapse
|
287
|
Abstract
Cholesterol-rich, apolipoprotein B (apoB)-containing lipoproteins are now widely accepted as the most important causal agents of atherosclerotic cardiovascular disease. Multiple unequivocal and orthogonal lines of evidence all converge on low-density lipoprotein and related particles as being the principal actors in the genesis of atherosclerosis. Here, we review the fundamental role of atherogenic apoB-containing lipoproteins in cardiovascular disease and several other humoral and parietal factors that are required to initiate and maintain arterial degeneration. The biology of foam cells and their interactions with high-density lipoproteins, including cholesterol efflux, are also briefly reviewed.
Collapse
Affiliation(s)
- Michael D Shapiro
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Sergio Fazio
- Center for Preventive Cardiology, Knight Cardiovascular Institute, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| |
Collapse
|
288
|
Yoon JJ, Lee YJ, Han BH, Choi ES, Kho MC, Park JH, Ahn YM, Kim HY, Kang DG, Lee HS. Protective effect of betulinic acid on early atherosclerosis in diabetic apolipoprotein-E gene knockout mice. Eur J Pharmacol 2017; 796:224-232. [DOI: 10.1016/j.ejphar.2016.11.044] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 11/23/2016] [Accepted: 11/24/2016] [Indexed: 12/27/2022]
|
289
|
Ungprasert P, Crowson CS, Matteson EL. Risk of cardiovascular disease among patients with sarcoidosis: a population-based retrospective cohort study, 1976-2013. Eur Respir J 2017; 49:1601290. [PMID: 28182567 PMCID: PMC5564441 DOI: 10.1183/13993003.01290-2016] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 11/11/2016] [Indexed: 01/06/2023]
Abstract
A higher incidence of cardiovascular disease (CVD) has been observed in several chronic inflammatory diseases. However, data on sarcoidosis are limited.In this study, 345 patients with incident sarcoidosis in Olmsted County (Minnesota, USA) during 1976-2013 were identified based on comprehensive medical record review. 345 sex- and age-matched comparators were also identified from the same underlying population. Medical records were individually reviewed for CVD, including coronary artery disease, congestive heart failure, atrial fibrillation, cerebrovascular accident, transient ischaemic attack, peripheral arterial disease and abdominal aortic aneurysm. Cox proportional hazards models with adjustment for age, sex, calendar year and cardiovascular risk factors were used to compare the rate of development of CVD between cases and comparators.The prevalence of CVD before the index date was not significantly different between the two groups. Adjusting for age, sex and calendar year, the risk of incident CVD after the index date was significantly elevated among patients with sarcoidosis with an adjusted hazard ratio of 1.57 (95% CI 1.15-2.16). Adjustment for cardiovascular risk factors yielded an adjusted hazard ratio of 1.65 (95% CI 1.08-2.53). Significantly increased risk was also observed for several types of CVD, including coronary artery disease, congestive heart failure, atrial fibrillation and cerebrovascular accident.Increased incidence of CVD among patients with sarcoidosis was demonstrated in this population-based cohort, even after controlling for baseline traditional atherosclerotic risk factors.
Collapse
Affiliation(s)
- Patompong Ungprasert
- Division of Rheumatology, Dept of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Cynthia S Crowson
- Division of Rheumatology, Dept of Internal Medicine, Mayo Clinic, Rochester, MN, USA
- Division of Biomedical Statistics and Informatics, Dept of Health Science Research, Mayo Clinic, Rochester, MN, USA
| | - Eric L Matteson
- Division of Rheumatology, Dept of Internal Medicine, Mayo Clinic, Rochester, MN, USA
- Division of Epidemiology, Dept of Health Science Research, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
290
|
Cipriano AF, Sallee A, Tayoba M, Cortez Alcaraz MC, Lin A, Guan RG, Zhao ZY, Liu H. Cytocompatibility and early inflammatory response of human endothelial cells in direct culture with Mg-Zn-Sr alloys. Acta Biomater 2017; 48:499-520. [PMID: 27746360 PMCID: PMC5873597 DOI: 10.1016/j.actbio.2016.10.020] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 10/04/2016] [Accepted: 10/12/2016] [Indexed: 12/18/2022]
Abstract
Crystalline Mg-Zinc (Zn)-Strontium (Sr) ternary alloys consist of elements naturally present in the human body and provide attractive mechanical and biodegradable properties for a variety of biomedical applications. The first objective of this study was to investigate the degradation and cytocompatibility of four Mg-4Zn-xSr alloys (x=0.15, 0.5, 1.0, 1.5wt%; designated as ZSr41A, B, C, and D respectively) in the direct culture with human umbilical vein endothelial cells (HUVEC) in vitro. The second objective was to investigate, for the first time, the early-stage inflammatory response in cultured HUVECs as indicated by the induction of vascular cellular adhesion molecule-1 (VCAM-1). The results showed that the 24-h in vitro degradation of the ZSr41 alloys containing a β-phase with a Zn/Sr at% ratio ∼1.5 was significantly faster than the ZSr41 alloys with Zn/Sr at% ∼1. Additionally, the adhesion density of HUVECs in the direct culture but not in direct contact with the ZSr41 alloys for up to 24h was not adversely affected by the degradation of the alloys. Importantly, neither culture media supplemented with up to 27.6mM Mg2+ ions nor media intentionally adjusted up to alkaline pH 9 induced any detectable adverse effects on HUVEC responses. In contrast, the significantly higher, yet non-cytotoxic, Zn2+ ion concentration from the degradation of ZSr41D alloy was likely the cause for the initially higher VCAM-1 expression on cultured HUVECs. Lastly, analysis of the HUVEC-ZSr41 interface showed near-complete absence of cell adhesion directly on the sample surface, most likely caused by either a high local alkalinity, change in surface topography, and/or surface composition. The direct culture method used in this study was proposed as a valuable tool for studying the design aspects of Zn-containing Mg-based biomaterials in vitro, in order to engineer solutions to address current shortcomings of Mg alloys for vascular device applications. STATEMENT OF SIGNIFICANCE Magnesium (Mg) alloys specifically designed for biodegradable implant applications have been the focus of biomedical research since the early 2000s. Physicochemical properties of Mg alloys make these metallic biomaterials excellent candidates for temporary biodegradable implants in orthopedic and cardiovascular applications. As Mg alloys continue to be investigated for biomedical applications, it is necessary to understand whether Mg-based materials or the alloying elements have the intrinsic ability to direct an immune response to improve implant integration while avoiding cell-biomaterial interactions leading to chronic inflammation and/or foreign body reactions. The present study utilized the direct culture method to investigate for the first time the in vitro transient inflammatory activation of endothelial cells induced by the degradation products of Zn-containing Mg alloys.
Collapse
Affiliation(s)
- Aaron F Cipriano
- Department of Bioengineering, University of California, Riverside, CA 92521, USA; Materials Science & Engineering Program, University of California, Riverside, CA 92521, USA
| | - Amy Sallee
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| | - Myla Tayoba
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| | | | - Alan Lin
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| | - Ren-Guo Guan
- School of Materials and Metallurgy, Northeastern University, Shenyang 110004, China
| | - Zhan-Yong Zhao
- School of Materials and Metallurgy, Northeastern University, Shenyang 110004, China
| | - Huinan Liu
- Department of Bioengineering, University of California, Riverside, CA 92521, USA; Materials Science & Engineering Program, University of California, Riverside, CA 92521, USA; Stem Cell Center, University of California, Riverside, CA 92521, USA; Cell, Molecular and Developmental Biology Program, University of California, Riverside, CA 92521, USA.
| |
Collapse
|
291
|
Chan JMS, Cheung MSH, Gibbs RGJ, Bhakoo KK. MRI detection of endothelial cell inflammation using targeted superparamagnetic particles of iron oxide (SPIO). Clin Transl Med 2017; 6:1. [PMID: 28044245 PMCID: PMC5206220 DOI: 10.1186/s40169-016-0134-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 12/21/2016] [Indexed: 01/10/2023] Open
Abstract
Background There is currently no clinical imaging technique available to assess the degree of inflammation associated with atherosclerotic plaques. This study aims to develop targeted superparamagnetic particles of iron oxide (SPIO) as a magnetic resonance imaging (MRI) probe for detecting inflamed endothelial cells. Methods The in vitro study consists of the characterisation and detection of inflammatory markers on activated endothelial cells by immunocytochemistry and MRI using biotinylated anti-P-selectin and anti-VCAM-1 (vascular cell adhesion molecule 1) antibody and streptavidin conjugated SPIO. Results Established an in vitro cellular model of endothelial inflammation induced with TNF-α (tumor necrosis factor alpha). Inflammation of endothelial cells was confirmed with both immunocytochemistry and MRI. These results revealed both a temporal and dose dependent expression of the inflammatory markers, P-selectin and VCAM-1, on exposure to TNF-α. Conclusion This study has demonstrated the development of an in vitro model to characterise and detect inflamed endothelial cells by immunocytochemistry and MRI. This will allow the future development of contrast agents and protocols for imaging vascular inflammation in atherosclerosis. This work may form the basis for a translational study to provide clinicians with a novel tool for the in vivo assessment of atherosclerosis.
Collapse
Affiliation(s)
- Joyce M S Chan
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China. .,Regional Vascular Unit, St Mary's Hospital, Imperial College Healthcare NHS Trust, Imperial College London, London, UK. .,Translational Molecular Imaging Group, Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, Helios, 138667, Singapore.
| | - Maggie S H Cheung
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China
| | - Richard G J Gibbs
- Regional Vascular Unit, St Mary's Hospital, Imperial College Healthcare NHS Trust, Imperial College London, London, UK
| | - Kishore K Bhakoo
- Translational Molecular Imaging Group, Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, Helios, 138667, Singapore
| |
Collapse
|
292
|
Lozhkin A, Vendrov AE, Pan H, Wickline SA, Madamanchi NR, Runge MS. NADPH oxidase 4 regulates vascular inflammation in aging and atherosclerosis. J Mol Cell Cardiol 2017; 102:10-21. [PMID: 27986445 PMCID: PMC5625334 DOI: 10.1016/j.yjmcc.2016.12.004] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 12/08/2016] [Accepted: 12/12/2016] [Indexed: 01/10/2023]
Abstract
We recently reported that increased NADPH oxidase 4 (NOX4) expression and activity during aging results in enhanced cellular and mitochondrial oxidative stress, vascular inflammation, dysfunction, and atherosclerosis. The goal of the present study was to elucidate the molecular mechanism(s) for these effects and determine the importance of NOX4 modulation of proinflammatory gene expression in mouse vascular smooth muscle cells (VSMCs). A novel peptide-mediated siRNA transfection approach was used to inhibit Nox4 expression with minimal cellular toxicity. Using melittin-derived peptide p5RHH, we achieved significantly higher transfection efficiency (92% vs. 85% with Lipofectamine) and decreased toxicity (p<0.001 vs. Lipofectamine in MTT and p<0.0001 vs. Lipofectamine in LDH assays) in VSMCs. TGFβ1 significantly upregulates Nox4 mRNA (p<0.01) and protein (p<0.01) expression in VSMCs. p5RHH-mediated Nox4 siRNA transfection greatly attenuated TGFβ1-induced upregulation of Nox4 mRNA (p<0.01) and protein (p<0.0001) levels and decreased hydrogen peroxide production (p<0.0001). Expression of pro-inflammatory genes Ccl2, Ccl5, Il6, and Vcam1 was significantly upregulated in VSMCs in several settings cells isolated from aged vs. young wild-type mice, in atherosclerotic arteries of Apoe-/- mice, and atherosclerotic human carotid arteries and correlated with NOX4 expression. p5RHH-mediated Nox4 siRNA transfection significantly attenuated the expression of these pro-inflammatory genes in TGFβ1-treated mouse VSMCs, with the highest degree of inhibition in the expression of Il6. p5RHH peptide-mediated knockdown of TGFβ-activated kinase 1 (TAK1, also known as Map3k7), Jun, and Rela, but not Nfkb2, downregulated TGFβ1-induced Nox4 expression in VSMCs. Together, these data demonstrate that increased expression and activation of NOX4, which might result from increased TGFβ1 levels seen during aging, induces a proinflammatory phenotype in VSMCs, enhancing atherosclerosis.
Collapse
Affiliation(s)
- Andrey Lozhkin
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan, Ann Arbor 48109, MI, USA
| | - Aleksandr E Vendrov
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan, Ann Arbor 48109, MI, USA
| | - Hua Pan
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis 63110, MO, USA
| | - Samuel A Wickline
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis 63110, MO, USA
| | - Nageswara R Madamanchi
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan, Ann Arbor 48109, MI, USA
| | - Marschall S Runge
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan, Ann Arbor 48109, MI, USA.
| |
Collapse
|
293
|
Biocompatible and target specific hydrophobically modified glycol chitosan nanoparticles. Biointerphases 2016; 11:04B301. [PMID: 27126597 DOI: 10.1116/1.4948265] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cardiovascular disease is the leading cause of death in the United States. Atherosclerosis is a major cause for cardiovascular diseases. Drugs that treat atherosclerosis usually act nonspecifically. To enhance drug delivery specificity, the authors developed a hydrophobically modified glycol chitosan (HGC) nanoparticle that can specifically target activated endothelial cells. The biocompatibility of these nanoparticles toward red blood cells and platelets was evaluated through hemolysis, platelet activation, platelet thrombogenicity, and platelet aggregation assays. The biocompatibility of these nanoparticles toward vascular endothelial cells was evaluated by their effects on endothelial cell growth, metabolic activity, and activation. The results demonstrated that HGC nanoparticles did not cause hemolysis, or affect platelet activation, thrombogenicity, and aggregation capability in vitro. The nanoparticles did not impair vascular endothelial cell growth or metabolic activities in vitro, and did not cause cell activation either. When conjugated with intercellular adhesion molecular 1 antibodies, HGC nanoparticles showed a significantly increased targeting specificity toward activated endothelial cells. These results suggested that HGC nanoparticles are likely compatible toward red blood cells, platelets, and endothelial cells, and they can be potentially used to identify activated endothelial cells at atherosclerotic lesion areas within the vasculature, and deliver therapeutic drugs.
Collapse
|
294
|
Nicolaou A, Zhao Z, Northoff BH, Sass K, Herbst A, Kohlmaier A, Chalaris A, Wolfrum C, Weber C, Steffens S, Rose-John S, Teupser D, Holdt LM. Adam17 Deficiency Promotes Atherosclerosis by Enhanced TNFR2 Signaling in Mice. Arterioscler Thromb Vasc Biol 2016; 37:247-257. [PMID: 28062509 DOI: 10.1161/atvbaha.116.308682] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Accepted: 11/28/2016] [Indexed: 12/31/2022]
Abstract
OBJECTIVE ADAM17 (a disintegrin and metalloproteinase 17) is a sheddase releasing different types of membrane-bound proteins, including adhesion molecules, cytokines, and their receptors as well as inflammatory mediators. Because these substrates modulate important mechanisms of atherosclerosis, we hypothesized that ADAM17 might be involved in the pathogenesis of this frequent disease. APPROACH AND RESULTS Because Adam17-knockout mice are not viable, we studied the effect of Adam17 deficiency on atherosclerosis in Adam17 hypomorphic mice (Adam17ex/ex), which have low residual Adam17 expression. To induce atherosclerosis, mice were crossed onto the low-density lipoprotein receptor (Ldlr)-deficient background. We found that Adam17ex/ex.Ldlr-/- mice developed ≈1.5-fold larger atherosclerotic lesions, which contained more macrophages and vascular smooth muscle cells than wild-type littermate controls (Adam17wt/wt.Ldlr-/-). Reduced Adam17-mediated shedding led to significantly increased protein levels of membrane-resident TNFα (tumor necrosis factor) and TNFR2 (tumor necrosis factor receptor 2), resulting in a constitutive activation of TNFR2 signaling. At the same time, Adam17 deficiency promoted proatherosclerotic cellular functions, such as increased proliferation and reduced apoptosis in cultured macrophages and vascular smooth muscle cells and increased adhesion of macrophages to vascular endothelial cells. Because siRNA (small interfering RNA)-mediated knockdown of Tnfr2 rescued from aberrant proliferation and from misregulation of apoptosis in Adam17-depleted cells, our data indicate that TNFR2 is an important effector of ADAM17 in our mouse model. CONCLUSIONS Our results provide evidence for an atheroprotective role of ADAM17, which might be mediated by cleaving membrane-bound TNFα and TNFR2, thereby preventing overactivation of endogenous TNFR2 signaling in cells of the vasculature.
Collapse
Affiliation(s)
- Alexandros Nicolaou
- From the Institute of Laboratory Medicine (A.N., B.H.N., K.S., A.H., A.K., D.T., L.M.H.) and Institute for Cardiovascular Prevention (Z.Z., C.Weber, S.S.), Ludwig-Maximilians-University Munich, Germany; Institute of Biochemistry, Christian Albrechts University, Kiel, Germany (A.C., S.R.-J.); Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland (C.Wolfrum); and German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C. Weber, S.S.)
| | - Zhen Zhao
- From the Institute of Laboratory Medicine (A.N., B.H.N., K.S., A.H., A.K., D.T., L.M.H.) and Institute for Cardiovascular Prevention (Z.Z., C.Weber, S.S.), Ludwig-Maximilians-University Munich, Germany; Institute of Biochemistry, Christian Albrechts University, Kiel, Germany (A.C., S.R.-J.); Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland (C.Wolfrum); and German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C. Weber, S.S.)
| | - Bernd H Northoff
- From the Institute of Laboratory Medicine (A.N., B.H.N., K.S., A.H., A.K., D.T., L.M.H.) and Institute for Cardiovascular Prevention (Z.Z., C.Weber, S.S.), Ludwig-Maximilians-University Munich, Germany; Institute of Biochemistry, Christian Albrechts University, Kiel, Germany (A.C., S.R.-J.); Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland (C.Wolfrum); and German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C. Weber, S.S.)
| | - Kristina Sass
- From the Institute of Laboratory Medicine (A.N., B.H.N., K.S., A.H., A.K., D.T., L.M.H.) and Institute for Cardiovascular Prevention (Z.Z., C.Weber, S.S.), Ludwig-Maximilians-University Munich, Germany; Institute of Biochemistry, Christian Albrechts University, Kiel, Germany (A.C., S.R.-J.); Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland (C.Wolfrum); and German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C. Weber, S.S.)
| | - Andreas Herbst
- From the Institute of Laboratory Medicine (A.N., B.H.N., K.S., A.H., A.K., D.T., L.M.H.) and Institute for Cardiovascular Prevention (Z.Z., C.Weber, S.S.), Ludwig-Maximilians-University Munich, Germany; Institute of Biochemistry, Christian Albrechts University, Kiel, Germany (A.C., S.R.-J.); Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland (C.Wolfrum); and German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C. Weber, S.S.)
| | - Alexander Kohlmaier
- From the Institute of Laboratory Medicine (A.N., B.H.N., K.S., A.H., A.K., D.T., L.M.H.) and Institute for Cardiovascular Prevention (Z.Z., C.Weber, S.S.), Ludwig-Maximilians-University Munich, Germany; Institute of Biochemistry, Christian Albrechts University, Kiel, Germany (A.C., S.R.-J.); Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland (C.Wolfrum); and German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C. Weber, S.S.)
| | - Athena Chalaris
- From the Institute of Laboratory Medicine (A.N., B.H.N., K.S., A.H., A.K., D.T., L.M.H.) and Institute for Cardiovascular Prevention (Z.Z., C.Weber, S.S.), Ludwig-Maximilians-University Munich, Germany; Institute of Biochemistry, Christian Albrechts University, Kiel, Germany (A.C., S.R.-J.); Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland (C.Wolfrum); and German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C. Weber, S.S.)
| | - Christian Wolfrum
- From the Institute of Laboratory Medicine (A.N., B.H.N., K.S., A.H., A.K., D.T., L.M.H.) and Institute for Cardiovascular Prevention (Z.Z., C.Weber, S.S.), Ludwig-Maximilians-University Munich, Germany; Institute of Biochemistry, Christian Albrechts University, Kiel, Germany (A.C., S.R.-J.); Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland (C.Wolfrum); and German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C. Weber, S.S.)
| | - Christian Weber
- From the Institute of Laboratory Medicine (A.N., B.H.N., K.S., A.H., A.K., D.T., L.M.H.) and Institute for Cardiovascular Prevention (Z.Z., C.Weber, S.S.), Ludwig-Maximilians-University Munich, Germany; Institute of Biochemistry, Christian Albrechts University, Kiel, Germany (A.C., S.R.-J.); Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland (C.Wolfrum); and German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C. Weber, S.S.)
| | - Sabine Steffens
- From the Institute of Laboratory Medicine (A.N., B.H.N., K.S., A.H., A.K., D.T., L.M.H.) and Institute for Cardiovascular Prevention (Z.Z., C.Weber, S.S.), Ludwig-Maximilians-University Munich, Germany; Institute of Biochemistry, Christian Albrechts University, Kiel, Germany (A.C., S.R.-J.); Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland (C.Wolfrum); and German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C. Weber, S.S.)
| | - Stefan Rose-John
- From the Institute of Laboratory Medicine (A.N., B.H.N., K.S., A.H., A.K., D.T., L.M.H.) and Institute for Cardiovascular Prevention (Z.Z., C.Weber, S.S.), Ludwig-Maximilians-University Munich, Germany; Institute of Biochemistry, Christian Albrechts University, Kiel, Germany (A.C., S.R.-J.); Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland (C.Wolfrum); and German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C. Weber, S.S.)
| | - Daniel Teupser
- From the Institute of Laboratory Medicine (A.N., B.H.N., K.S., A.H., A.K., D.T., L.M.H.) and Institute for Cardiovascular Prevention (Z.Z., C.Weber, S.S.), Ludwig-Maximilians-University Munich, Germany; Institute of Biochemistry, Christian Albrechts University, Kiel, Germany (A.C., S.R.-J.); Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland (C.Wolfrum); and German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C. Weber, S.S.)
| | - Lesca M Holdt
- From the Institute of Laboratory Medicine (A.N., B.H.N., K.S., A.H., A.K., D.T., L.M.H.) and Institute for Cardiovascular Prevention (Z.Z., C.Weber, S.S.), Ludwig-Maximilians-University Munich, Germany; Institute of Biochemistry, Christian Albrechts University, Kiel, Germany (A.C., S.R.-J.); Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland (C.Wolfrum); and German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany (C. Weber, S.S.).
| |
Collapse
|
295
|
Soltani R, Hashemi M, Farazmand A, Asghari G, Heshmat-Ghahdarijani K, Kharazmkia A, Ghanadian SM. Evaluation of the Effects ofCucumis sativusSeed Extract on Serum Lipids in Adult Hyperlipidemic Patients: A Randomized Double-Blind Placebo-Controlled Clinical Trial. J Food Sci 2016; 82:214-218. [DOI: 10.1111/1750-3841.13569] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 10/12/2016] [Accepted: 11/01/2016] [Indexed: 12/30/2022]
Affiliation(s)
- Rasool Soltani
- Dept. of Clinical Pharmacy and Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences; Isfahan Univ. of Medical Sciences; Isfahan Iran
| | - Mohammad Hashemi
- Dept. of Cardiology, School of Medicine; Isfahan Univ. of Medical Sciences; Isfahan Iran
| | - Alimohammad Farazmand
- Dept. of Clinical Pharmacy and Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences; Isfahan Univ. of Medical Sciences; Isfahan Iran
| | - Gholamreza Asghari
- Dept. of Pharmacognosy, School of Pharmacy and Pharmaceutical Sciences; Isfahan Univ. of Medical Sciences; Isfahan Iran
| | | | - Ali Kharazmkia
- Dept. of Clinical Pharmacy, School of Pharmacy; Lorestan Univ. of Medical Sciences; Khorramabad Iran
| | - Syed Mustafa Ghanadian
- Dept. of Pharmacognosy, School of Pharmacy and Pharmaceutical Sciences; Isfahan Univ. of Medical Sciences; Isfahan Iran
| |
Collapse
|
296
|
Ruiz M, Frej C, Holmér A, Guo LJ, Tran S, Dahlbäck B. High-Density Lipoprotein-Associated Apolipoprotein M Limits Endothelial Inflammation by Delivering Sphingosine-1-Phosphate to the Sphingosine-1-Phosphate Receptor 1. Arterioscler Thromb Vasc Biol 2016; 37:118-129. [PMID: 27879252 DOI: 10.1161/atvbaha.116.308435] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 11/08/2016] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Plasma high-density lipoproteins (HDL) are potent antiatherogenic and anti-inflammatory particles. However, HDL particles are highly heterogenic in composition, and different HDL-mediated functions can be ascribed to different subclasses of HDL. Only a small HDL population contains apolipoprotein M (ApoM), which is the main plasma carrier of the bioactive lipid mediator sphingosine-1-phosphate (S1P). Vascular inflammation is modulated by S1P, but both pro- and anti-inflammatory roles have been ascribed to S1P. The goal of this study is to elucidate the role of ApoM and S1P in endothelial anti-inflammatory events related to HDL. APPROACH AND RESULTS Aortic or brain human primary endothelial cells were challenged with tumor necrosis factor-α (TNF-α) as inflammatory stimuli. The presence of recombinant ApoM-bound S1P or ApoM-containing HDL reduced the abundance of adhesion molecules in the cell surface, whereas ApoM and ApoM-lacking HDL did not. Specifically, ApoM-bound S1P decreased vascular adhesion molecule-1 (VCAM-1) and E-selectin surface abundance but not intercellular adhesion molecule-1. Albumin, which is an alternative S1P carrier, was less efficient in inhibiting VCAM-1 than ApoM-bound S1P. The activation of the S1P receptor 1 was sufficient and required to promote anti-inflammation. Moreover, ApoM-bound S1P induced the rearrangement of the expression of S1P-related genes to counteract TNF-α. Functionally, HDL/ApoM/S1P limited monocyte adhesion to the endothelium and maintained endothelial barrier integrity under inflammatory conditions. CONCLUSIONS ApoM-bound S1P is a key component of HDL and is responsible for several HDL-associated protective functions in the endothelium, including regulation of adhesion molecule abundance, leukocyte-endothelial adhesion, and endothelial barrier.
Collapse
Affiliation(s)
- Mario Ruiz
- From the Department of Translational Medicine, Skåne University Hospital, Lund University, Malmö, Sweden.
| | - Cecilia Frej
- From the Department of Translational Medicine, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Andreas Holmér
- From the Department of Translational Medicine, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Li J Guo
- From the Department of Translational Medicine, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Sinh Tran
- From the Department of Translational Medicine, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Björn Dahlbäck
- From the Department of Translational Medicine, Skåne University Hospital, Lund University, Malmö, Sweden
| |
Collapse
|
297
|
Chen X, Qian S, Hoggatt A, Tang H, Hacker TA, Obukhov AG, Herring PB, Seye CI. Endothelial Cell-Specific Deletion of P2Y2 Receptor Promotes Plaque Stability in Atherosclerosis-Susceptible ApoE-Null Mice. Arterioscler Thromb Vasc Biol 2016; 37:75-83. [PMID: 27856454 DOI: 10.1161/atvbaha.116.308561] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 11/01/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Nucleotide P2Y2 receptor (P2Y2R) contributes to vascular inflammation by increasing vascular cell adhesion molecule-1 expression in endothelial cells (EC), and global P2Y2R deficiency prevents fatty streak formation in apolipoprotein E null (ApoE-/-) mice. Because P2Y2R is ubiquitously expressed in vascular cells, we investigated the contribution of endothelial P2Y2R in the pathogenesis of atherosclerosis. APPROACH AND RESULTS EC-specific P2Y2R-deficient mice were generated by breeding VEcadherin5-Cre mice with the P2Y2R floxed mice. Endothelial P2Y2R deficiency reduced endothelial nitric oxide synthase activity and significantly altered ATP- and UTP (uridine 5'-triphosphate)-induced vasorelaxation without affecting vasodilatory responses to acetylcholine. Telemetric blood pressure and echocardiography measurements indicated that EC-specific P2Y2R-deficient mice did not develop hypertension. We investigated the role of endothelial P2Y2R in the development of atherosclerotic lesions by crossing the EC-specific P2Y2R knockout mice onto an ApoE-/- background and evaluated lesion development after feeding a standard chow diet for 25 weeks. Histopathologic examination demonstrated reduced atherosclerotic lesions in the aortic sinus and entire aorta, decreased macrophage infiltration, and increased smooth muscle cell and collagen content, leading to the formation of a subendothelial fibrous cap in EC-specific P2Y2R-deficient ApoE-/- mice. Expression and proteolytic activity of matrix metalloproteinase-2 was significantly reduced in atherosclerotic lesions from EC-specific P2Y2R-deficient ApoE-/- mice. Furthermore, EC-specific P2Y2R deficiency inhibited nitric oxide production, leading to significant increase in smooth muscle cell migration out of aortic explants. CONCLUSIONS EC-specific P2Y2R deficiency reduces atherosclerotic burden and promotes plaque stability in ApoE-/- mice through impaired macrophage infiltration acting together with reduced matrix metalloproteinase-2 activity and increased smooth muscle cell migration.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Aorta, Thoracic/physiopathology
- Aortic Diseases/genetics
- Aortic Diseases/metabolism
- Aortic Diseases/pathology
- Aortic Diseases/prevention & control
- Apolipoproteins E/deficiency
- Apolipoproteins E/genetics
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/prevention & control
- Cell Movement
- Cells, Cultured
- Collagen/metabolism
- Disease Models, Animal
- Disease Progression
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Fibrosis
- Macrophages/metabolism
- Macrophages/pathology
- Male
- Matrix Metalloproteinase 2/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Nitric Oxide/metabolism
- Nitric Oxide Synthase Type III/metabolism
- Plaque, Atherosclerotic
- Purinergic P2Y Receptor Agonists/pharmacology
- Receptors, Purinergic P2Y2/deficiency
- Receptors, Purinergic P2Y2/genetics
- Rupture, Spontaneous
- Signal Transduction
- Vasodilation/drug effects
- Vasodilator Agents/pharmacology
Collapse
Affiliation(s)
- Xingjuan Chen
- From the Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis (X.C., S.Q., A.H., H.T., A.G.O., P.B.H., C.I.S.); and Department of Medicine, University of Wisconsin-Madison (T.A.H.)
| | - Shaomin Qian
- From the Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis (X.C., S.Q., A.H., H.T., A.G.O., P.B.H., C.I.S.); and Department of Medicine, University of Wisconsin-Madison (T.A.H.)
| | - April Hoggatt
- From the Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis (X.C., S.Q., A.H., H.T., A.G.O., P.B.H., C.I.S.); and Department of Medicine, University of Wisconsin-Madison (T.A.H.)
| | - Hongying Tang
- From the Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis (X.C., S.Q., A.H., H.T., A.G.O., P.B.H., C.I.S.); and Department of Medicine, University of Wisconsin-Madison (T.A.H.)
| | - Timothy A Hacker
- From the Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis (X.C., S.Q., A.H., H.T., A.G.O., P.B.H., C.I.S.); and Department of Medicine, University of Wisconsin-Madison (T.A.H.)
| | - Alexander G Obukhov
- From the Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis (X.C., S.Q., A.H., H.T., A.G.O., P.B.H., C.I.S.); and Department of Medicine, University of Wisconsin-Madison (T.A.H.)
| | - Paul B Herring
- From the Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis (X.C., S.Q., A.H., H.T., A.G.O., P.B.H., C.I.S.); and Department of Medicine, University of Wisconsin-Madison (T.A.H.)
| | - Cheikh I Seye
- From the Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis (X.C., S.Q., A.H., H.T., A.G.O., P.B.H., C.I.S.); and Department of Medicine, University of Wisconsin-Madison (T.A.H.).
| |
Collapse
|
298
|
Yuan X, Chen J, Dai M. Paeonol promotes microRNA-126 expression to inhibit monocyte adhesion to ox-LDL-injured vascular endothelial cells and block the activation of the PI3K/Akt/NF-κB pathway. Int J Mol Med 2016; 38:1871-1878. [PMID: 27748840 DOI: 10.3892/ijmm.2016.2778] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 10/03/2016] [Indexed: 11/06/2022] Open
Abstract
Paeonol (2'-hydroxy-4'-methoxyacetophenone) is an active component isolated from the root of Paeonia Suffruticosa Andrews. We previously found that paeonol inhibited vascular cell adhesion molecule-1 (VCAM‑1) expression, and thus may be useful for the prevention and treatment of rabbit atherosclerosis (AS); however, the underlying mechanisms are not yet well known. Recently, microRNAs (miRNAs or miRs) have been reported to play an important role in the pathogenesis of AS. In the present study, we examined the effects of paeonol on miRNA-126 (miR‑126) expression, and its ability to inhibit monocyte adhesion to oxidized low-density lipoprotein (ox-LDL)-injured vascular endothelial cells (VECs). VECs were isolated from the rat thoracic aorta and stimulated with ox-LDL (20 mg/l) in the presence of paeonol. We found that miR‑126 had a lower expression in the ox-LDL-injured VECs, and VCAM‑1 was identified as a target gene of miR‑126. Furthermore, paeonol promoted miR‑126 expression and suppressed VCAM‑1 expression at the mRNA and protein level. It also inhibited monocyte adhesion to ox-LDL-injured VECs through the promotion of miR‑126 expression. Furthermore, it was demonstrated that paeonol blocked the activation of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/nuclear factor-κB (NF-κB) signaling pathway by promoting miR-126 expression. Taken together, and to the best of our knowledge, the findings of this study provide the first evidence that paeonol promotes miR‑126 expression to inhibit monocyte adhesion to ox-LDL-injured VECs and block the activation of the PI3K/Akt/NF-κB signaling pathway. Our data suggest that miR‑126 plays a crucial role in vascular inflammation and may be an important therapeutic target in the treatment of AS with the use of paeonol.
Collapse
Affiliation(s)
- Xusheng Yuan
- Key Laboratory of Chinese Medicine Research and Development, Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230038, P.R. China
| | - Junjun Chen
- Key Laboratory of Xin'an Medicine, Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230038, P.R. China
| | - Min Dai
- Key Laboratory of Chinese Medicine Research and Development, Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230038, P.R. China
| |
Collapse
|
299
|
Kirichenko TV, Sobenin IA, Nikolic D, Rizzo M, Orekhov AN. Anti-cytokine therapy for prevention of atherosclerosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2016; 23:1198-1210. [PMID: 26781385 DOI: 10.1016/j.phymed.2015.12.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 12/01/2015] [Accepted: 12/04/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND Currently a chronic inflammation is considered to be the one of the most important reasons of the atherosclerosis progression. A huge amount of researches over the past few decades are devoted to study the various mechanisms of inflammation in the development of atherosclerotic lesions. PURPOSE To review current capabilities of anti-inflammatory therapy for the prevention and treatment of atherosclerosis and its clinical manifestations. METHODS Appropriate articles on inflammatory cytokines in atherosclerosis and anti-inflammatory prevention of atherosclerosis were searched in PubMed Database from their respective inceptions until October 2015. SECTIONS "The role of inflammatory cytokines in the development of atherosclerotic lesions" describes available data on the possible inflammatory mechanisms of the atherogenesis with a special attention to the role of cytokines. "Modern experience of anti-inflammatory therapy for the treatment of atherosclerosis" describes modern anti-inflammatory preparations with anti-atherosclerotic effect including natural preparations. In "the development of anti-inflammatory herbal preparation for atherosclerosis prevention" an algorithm is demonstrated that includes screening of anti-cytokine activity of different natural products, the development of the most effective combination and estimation of its effect in cell culture model, in animal model of the acute aseptic inflammation and in a pilot clinical trial. A natural preparation "Inflaminat" based on black elder berries (Sambucus nigra L.), violet tricolor herb (Viola tricolor L.) and calendula flowers (Calendula officinalis L.) possessing anti-cytokine activity was developed using the designed algorithm. The results of the following 2-year double blind placebo-controlled clinical study show that "Inflaminat" reduces carotid IMT progression, i.e. has anti-atherosclerotic effect. CONCLUSION Anti-cytokine therapy may be a promising direction in moderation of atherogenesis, especially when it begins on the early stages of subclinical atherosclerosis. The use of herbal preparations with anti-cytokine mechanism of action is the most perspective for timely prevention of atherosclerosis, as they have no significant side effects and can be prescribed for long-term administration.
Collapse
Affiliation(s)
- Tatiana V Kirichenko
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, 4-1-207, Osennaya Str., 121609, Moscow, Russia.
| | - Igor A Sobenin
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Str., 125315, Moscow, Russia; Russian Cardiology Research and Production Complex, 15-a 3rd Cherepkovskaya Str., 121552, Moscow, Russia
| | - Dragana Nikolic
- Biomedical Department of Internal Medicine and Medical Specialties, University of Palermo, 141 Via del Vespro, 90127, Palermo, Italy
| | - Manfredi Rizzo
- Biomedical Department of Internal Medicine and Medical Specialties, University of Palermo, 141 Via del Vespro, 90127, Palermo, Italy; Euro-Mediterranean Institute of Science and Technology, Via Emerico Amari 123, 90139, Palermo, Italy
| | - Alexander N Orekhov
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, 4-1-207, Osennaya Str., 121609, Moscow, Russia; Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Str., 125315, Moscow, Russia
| |
Collapse
|
300
|
Herbin O, Regelmann AG, Ramkhelawon B, Weinstein EG, Moore KJ, Alexandropoulos K. Monocyte Adhesion and Plaque Recruitment During Atherosclerosis Development Is Regulated by the Adapter Protein Chat-H/SHEP1. Arterioscler Thromb Vasc Biol 2016; 36:1791-801. [PMID: 27417580 PMCID: PMC5001917 DOI: 10.1161/atvbaha.116.308014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 07/04/2016] [Indexed: 01/01/2023]
Abstract
OBJECTIVE The chronic inflammation associated with atherosclerosis is caused by lipid deposition followed by leukocyte recruitment to the arterial wall. We previously showed that the hematopoietic cell-specific adaptor protein Cas- and Hef1-associated signal transducer hematopoietic isoform (Chat-H)/SHEP1 regulated lymphocyte adhesion and migration. In this study, we analyzed the role of Chat-H in atherosclerosis development. APPROACH AND RESULTS Using Chat-H-deficient bone marrow transplantation in low-density lipoprotein receptor-deficient mice, we found that Chat-H regulated atherosclerotic plaque formation. Chat-H deficiency in hematopoietic cells associated with lower plaque complexity and fewer leukocytes in the lesions, whereas myeloid-specific deletion of Chat-H was sufficient for conferring atheroprotection. Chat-H deficiency resulted in reduced recruitment of classical Ly6c(high) and nonclassical Ly6c(low) monocytes to the plaques, which was accompanied by increased numbers of both monocyte subsets in the blood. This associated with defective adhesion of Chat-H-deficient Ly6c(high) and Ly6c(low) monocytes to vascular cell adhesion molecule-1 in vitro and impaired infiltration of fluorescent bead-loaded monocytes to atherosclerotic plaques. In contrast, Chat-H was dispensable for CX3CL1 and CCR1/CCR5-dependent migration of monocytes. CONCLUSIONS Our findings highlight Chat-H as a key protein that regulates atherosclerosis development by controlling monocyte adhesion and recruitment to the plaques and identify a novel target that may be exploited for treating atherosclerosis.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/deficiency
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Antigens, Ly/metabolism
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/prevention & control
- Bone Marrow Transplantation
- Cell Adhesion
- Cells, Cultured
- Chemotaxis, Leukocyte
- Disease Models, Animal
- Genotype
- Macrophages/metabolism
- Macrophages/pathology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Monocytes/metabolism
- Monocytes/pathology
- Neutrophils/metabolism
- Neutrophils/pathology
- Phenotype
- Plaque, Atherosclerotic
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
- Signal Transduction
- Vascular Cell Adhesion Molecule-1/metabolism
Collapse
Affiliation(s)
- Olivier Herbin
- From the Icahn School of Medicine at Mount Sinai, Department of Medicine, The Immunology Institute, New York (O.H., E.G.W., K.A.); Quartzy, Inc, Palo Alto, CA (A.G.R.); and Leon H. Charney Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (B.R., K.J.M.)
| | - Adam G Regelmann
- From the Icahn School of Medicine at Mount Sinai, Department of Medicine, The Immunology Institute, New York (O.H., E.G.W., K.A.); Quartzy, Inc, Palo Alto, CA (A.G.R.); and Leon H. Charney Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (B.R., K.J.M.)
| | - Bhama Ramkhelawon
- From the Icahn School of Medicine at Mount Sinai, Department of Medicine, The Immunology Institute, New York (O.H., E.G.W., K.A.); Quartzy, Inc, Palo Alto, CA (A.G.R.); and Leon H. Charney Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (B.R., K.J.M.)
| | - Erica G Weinstein
- From the Icahn School of Medicine at Mount Sinai, Department of Medicine, The Immunology Institute, New York (O.H., E.G.W., K.A.); Quartzy, Inc, Palo Alto, CA (A.G.R.); and Leon H. Charney Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (B.R., K.J.M.)
| | - Kathryn J Moore
- From the Icahn School of Medicine at Mount Sinai, Department of Medicine, The Immunology Institute, New York (O.H., E.G.W., K.A.); Quartzy, Inc, Palo Alto, CA (A.G.R.); and Leon H. Charney Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (B.R., K.J.M.)
| | - Konstantina Alexandropoulos
- From the Icahn School of Medicine at Mount Sinai, Department of Medicine, The Immunology Institute, New York (O.H., E.G.W., K.A.); Quartzy, Inc, Palo Alto, CA (A.G.R.); and Leon H. Charney Division of Cardiology, Department of Medicine, NYU School of Medicine, New York (B.R., K.J.M.).
| |
Collapse
|