1
|
Wang Y, Li G, Chen B, Shakir G, Volz M, van der Vorst EPC, Maas SL, Geiger M, Jethwa C, Bartelt A, Li Z, Wettich J, Sachs N, Maegdefessel L, Nazari Jahantigh M, Hristov M, Lacy M, Lutz B, Weber C, Herzig S, Guillamat Prats R, Steffens S. Myeloid cannabinoid CB1 receptor deletion confers atheroprotection in male mice by reducing macrophage proliferation in a sex-dependent manner. Cardiovasc Res 2024; 120:1411-1426. [PMID: 38838211 PMCID: PMC11481387 DOI: 10.1093/cvr/cvae125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/21/2024] [Accepted: 05/01/2024] [Indexed: 06/07/2024] Open
Abstract
AIMS Although the cannabinoid CB1 receptor has been implicated in atherosclerosis, its cell-specific effects in this disease are not well understood. To address this, we generated a transgenic mouse model to study the role of myeloid CB1 signalling in atherosclerosis. METHODS AND RESULTS Here, we report that male mice with myeloid-specific Cnr1 deficiency on atherogenic background developed smaller lesions and necrotic cores than controls, while only minor genotype differences were observed in females. Male Cnr1-deficient mice showed reduced arterial monocyte recruitment and macrophage proliferation with less inflammatory phenotype. The sex-specific differences in proliferation were dependent on oestrogen receptor (ER)α-oestradiol signalling. Kinase activity profiling identified a CB1-dependent regulation of p53 and cyclin-dependent kinases. Transcriptomic profiling further revealed chromatin modifications, mRNA processing, and mitochondrial respiration among the key processes affected by CB1 signalling, which was supported by metabolic flux assays. Chronic administration of the peripherally restricted CB1 antagonist JD5037 inhibited plaque progression and macrophage proliferation, but only in male mice. Finally, CNR1 expression was detectable in human carotid endarterectomy plaques and inversely correlated with proliferation, oxidative metabolism, and inflammatory markers, suggesting a possible implication of CB1-dependent regulation in human pathophysiology. CONCLUSION Impaired macrophage CB1 signalling is atheroprotective by limiting their arterial recruitment, proliferation, and inflammatory reprogramming in male mice. The importance of macrophage CB1 signalling appears to be sex-dependent.
Collapse
Affiliation(s)
- Yong Wang
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität Munich, Pettenkoferstr. 9, 80336 Munich, Germany
| | - Guo Li
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität Munich, Pettenkoferstr. 9, 80336 Munich, Germany
| | - Bingni Chen
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität Munich, Pettenkoferstr. 9, 80336 Munich, Germany
| | - George Shakir
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität Munich, Pettenkoferstr. 9, 80336 Munich, Germany
| | - Mario Volz
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität Munich, Pettenkoferstr. 9, 80336 Munich, Germany
| | - Emiel P C van der Vorst
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität Munich, Pettenkoferstr. 9, 80336 Munich, Germany
- Institute for Molecular Cardiovascular Research (IMCAR), Aachen-Maastricht Institute for CardioRenal Disease (AMICARE) and Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany
| | - Sanne L Maas
- Institute for Molecular Cardiovascular Research (IMCAR), Aachen-Maastricht Institute for CardioRenal Disease (AMICARE) and Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany
| | - Martina Geiger
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität Munich, Pettenkoferstr. 9, 80336 Munich, Germany
| | - Carolin Jethwa
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität Munich, Pettenkoferstr. 9, 80336 Munich, Germany
| | - Alexander Bartelt
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität Munich, Pettenkoferstr. 9, 80336 Munich, Germany
- DZHK (German Center for Cardiovasular Research), partner site Munich Heart Alliance, Pettenkoferstr. 9, 80336 Munich, Germany
- Institute for Diabetes and Cancer, Helmholtz Zentrum Munich, Neuherberg, Germany
- Department of Molecular Metabolism & Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Boston, USA
| | - Zhaolong Li
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar—Technical University Munich (TUM), Munich, Germany
| | - Justus Wettich
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar—Technical University Munich (TUM), Munich, Germany
| | - Nadja Sachs
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar—Technical University Munich (TUM), Munich, Germany
| | - Lars Maegdefessel
- DZHK (German Center for Cardiovasular Research), partner site Munich Heart Alliance, Pettenkoferstr. 9, 80336 Munich, Germany
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar—Technical University Munich (TUM), Munich, Germany
| | - Maliheh Nazari Jahantigh
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität Munich, Pettenkoferstr. 9, 80336 Munich, Germany
| | - Michael Hristov
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität Munich, Pettenkoferstr. 9, 80336 Munich, Germany
| | - Michael Lacy
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität Munich, Pettenkoferstr. 9, 80336 Munich, Germany
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center, Mainz, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität Munich, Pettenkoferstr. 9, 80336 Munich, Germany
- DZHK (German Center for Cardiovasular Research), partner site Munich Heart Alliance, Pettenkoferstr. 9, 80336 Munich, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
- Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Straße 17, 81377 Munich, Germany
| | - Stephan Herzig
- DZHK (German Center for Cardiovasular Research), partner site Munich Heart Alliance, Pettenkoferstr. 9, 80336 Munich, Germany
- Institute for Diabetes and Cancer, Helmholtz Zentrum Munich, Neuherberg, Germany
- Chair Molecular Metabolic Control, TU Munich, Ismaninger Str. 22, 81675 Munich, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany
| | - Raquel Guillamat Prats
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität Munich, Pettenkoferstr. 9, 80336 Munich, Germany
| | - Sabine Steffens
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität Munich, Pettenkoferstr. 9, 80336 Munich, Germany
- DZHK (German Center for Cardiovasular Research), partner site Munich Heart Alliance, Pettenkoferstr. 9, 80336 Munich, Germany
| |
Collapse
|
2
|
Guillamat-Prats R, Hering D, Derle A, Rami M, Härdtner C, Santovito D, Rinne P, Bindila L, Hristov M, Pagano S, Vuilleumier N, Schmid S, Janjic A, Enard W, Weber C, Maegdefessel L, Faussner A, Hilgendorf I, Steffens S. GPR55 in B cells limits atherosclerosis development and regulates plasma cell maturation. NATURE CARDIOVASCULAR RESEARCH 2022; 1:1056-1071. [PMID: 36523570 PMCID: PMC7613934 DOI: 10.1038/s44161-022-00155-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 09/27/2022] [Indexed: 06/17/2023]
Abstract
Dissecting the pathways regulating the adaptive immune response in atherosclerosis is of particular therapeutic interest. Here we report that the lipid G-protein coupled receptor GPR55 is highly expressed by splenic plasma cells (PC), upregulated in mouse spleens during atherogenesis and human unstable or ruptured compared to stable plaques. Gpr55-deficient mice developed larger atherosclerotic plaques with increased necrotic core size compared to their corresponding controls. Lack of GPR55 hyperactivated B cells, disturbed PC maturation and resulted in immunoglobulin (Ig)G overproduction. B cell-specific Gpr55 depletion or adoptive transfer of Gpr55-deficient B cells was sufficient to promote plaque development and elevated IgG titers. In vitro, the endogenous GPR55 ligand lysophsophatidylinositol (LPI) enhanced PC proliferation, whereas GPR55 antagonism blocked PC maturation and increased their mitochondrial content. Collectively, these discoveries provide previously undefined evidence for GPR55 in B cells as a key modulator of the adaptive immune response in atherosclerosis.
Collapse
Affiliation(s)
- Raquel Guillamat-Prats
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Daniel Hering
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Abhishek Derle
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Martina Rami
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Carmen Härdtner
- Department of Cardiology and Angiology I, Heart Center and Faculty of Medicine, University of Freiburg. Freiburg, Germany
| | - Donato Santovito
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (MHA), Munich, Germany
- Institute for Genetic and Biomedical Research (IRGB), Unit of Milan, National Research Council, Milan, Italy
| | - Petteri Rinne
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Laura Bindila
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Michael Hristov
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Sabrina Pagano
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals and Faculty of Medicine
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals and Faculty of Medicine
| | - Sofie Schmid
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar - Technical University Munich (TUM), Munich, Germany
| | - Aleksandar Janjic
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians University, Martinsried, Germany
| | - Wolfgang Enard
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians University, Martinsried, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (MHA), Munich, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Lars Maegdefessel
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (MHA), Munich, Germany
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar - Technical University Munich (TUM), Munich, Germany
| | - Alexander Faussner
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Ingo Hilgendorf
- Department of Cardiology and Angiology I, Heart Center and Faculty of Medicine, University of Freiburg. Freiburg, Germany
- Institute for Experimental Cardiovascular Medicine, Heart Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sabine Steffens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (MHA), Munich, Germany
| |
Collapse
|
3
|
Kong P, Cui ZY, Huang XF, Zhang DD, Guo RJ, Han M. Inflammation and atherosclerosis: signaling pathways and therapeutic intervention. Signal Transduct Target Ther 2022; 7:131. [PMID: 35459215 PMCID: PMC9033871 DOI: 10.1038/s41392-022-00955-7] [Citation(s) in RCA: 464] [Impact Index Per Article: 154.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 02/08/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory vascular disease driven by traditional and nontraditional risk factors. Genome-wide association combined with clonal lineage tracing and clinical trials have demonstrated that innate and adaptive immune responses can promote or quell atherosclerosis. Several signaling pathways, that are associated with the inflammatory response, have been implicated within atherosclerosis such as NLRP3 inflammasome, toll-like receptors, proprotein convertase subtilisin/kexin type 9, Notch and Wnt signaling pathways, which are of importance for atherosclerosis development and regression. Targeting inflammatory pathways, especially the NLRP3 inflammasome pathway and its regulated inflammatory cytokine interleukin-1β, could represent an attractive new route for the treatment of atherosclerotic diseases. Herein, we summarize the knowledge on cellular participants and key inflammatory signaling pathways in atherosclerosis, and discuss the preclinical studies targeting these key pathways for atherosclerosis, the clinical trials that are going to target some of these processes, and the effects of quelling inflammation and atherosclerosis in the clinic.
Collapse
Affiliation(s)
- Peng Kong
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Zi-Yang Cui
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Xiao-Fu Huang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Dan-Dan Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Rui-Juan Guo
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Mei Han
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China.
| |
Collapse
|
4
|
Sakic A, Chaabane C, Ambartsumian N, Klingelhöfer J, Lemeille S, Kwak BR, Grigorian M, Bochaton-Piallat ML. Neutralization of S100A4 induces stabilization of atherosclerotic plaques: role of smooth muscle cells. Cardiovasc Res 2022; 118:141-155. [PMID: 33135065 PMCID: PMC8752361 DOI: 10.1093/cvr/cvaa311] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 10/20/2020] [Indexed: 01/20/2023] Open
Abstract
AIMS During atherosclerosis, smooth muscle cells (SMCs) accumulate in the intima where they switch from a contractile to a synthetic phenotype. From porcine coronary artery, we isolated spindle-shaped (S) SMCs exhibiting features of the contractile phenotype and rhomboid (R) SMCs typical of the synthetic phenotype. S100A4 was identified as a marker of R-SMCs in vitro and intimal SMCs, in pig and man. S100A4 exhibits intra- and extracellular functions. In this study, we investigated the role of extracellular S100A4 in SMC phenotypic transition. METHODS AND RESULTS S-SMCs were treated with oligomeric recombinant S100A4 (oS100A4), which induced nuclear factor (NF)-κB activation. Treatment of S-SMCs with oS100A4 in combination with platelet-derived growth factor (PDGF)-BB induced a complete SMC transition towards a pro-inflammatory R-phenotype associated with NF-κB activation, through toll-like receptor-4. RNA sequencing of cells treated with oS100A4/PDGF-BB revealed a strong up-regulation of pro-inflammatory genes and enrichment of transcription factor binding sites essential for SMC phenotypic transition. In a mouse model of established atherosclerosis, neutralization of extracellular S100A4 decreased area of atherosclerotic lesions, necrotic core, and CD68 expression and increased α-smooth muscle actin and smooth muscle myosin heavy chain expression. CONCLUSION We suggest that the neutralization of extracellular S100A4 promotes the stabilization of atherosclerotic plaques. Extracellular S100A4 could be a new target to influence the evolution of atherosclerotic plaques.
Collapse
MESH Headings
- Actins/metabolism
- Animals
- Antibodies, Neutralizing/pharmacology
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/metabolism
- Aorta/drug effects
- Aorta/metabolism
- Aorta/pathology
- Aortic Diseases/drug therapy
- Aortic Diseases/genetics
- Aortic Diseases/metabolism
- Aortic Diseases/pathology
- Atherosclerosis/drug therapy
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Becaplermin/pharmacology
- Cells, Cultured
- Disease Models, Animal
- Humans
- Mice, Inbred C57BL
- Mice, Knockout, ApoE
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Myosin Heavy Chains/metabolism
- Phenotype
- Plaque, Atherosclerotic
- S100 Calcium-Binding Protein A4/antagonists & inhibitors
- S100 Calcium-Binding Protein A4/metabolism
- S100 Calcium-Binding Protein A4/pharmacology
- Signal Transduction
- Smooth Muscle Myosins/metabolism
- Sus scrofa
- Toll-Like Receptor 4/metabolism
- Mice
Collapse
Affiliation(s)
- Antonija Sakic
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Chiraz Chaabane
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Noona Ambartsumian
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Jörg Klingelhöfer
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Sylvain Lemeille
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Brenda R Kwak
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Mariam Grigorian
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | | |
Collapse
|
5
|
Mohmmad‐Rezaei M, Arefnezhad R, Ahmadi R, Abdollahpour‐Alitappeh M, Mirzaei Y, Arjmand M, Ferns GA, Bashash D, Bagheri N. An overview of the innate and adaptive immune system in atherosclerosis. IUBMB Life 2021; 73:64-91. [DOI: 10.1002/iub.2425] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 11/24/2020] [Indexed: 12/13/2022]
Abstract
AbstractCardiovascular disease is the leading cause of death globally. Coronary artery disease (CAD) is a chronic inflammatory disease usually caused by atherosclerosis, in which the coronary arteries become narrowed by atheromatous plaque. Plaques in atherosclerosis are formed through the accumulation of lipids and various immune cells. Both adaptive and innate immune systems are involved in the pathogenesis of atherosclerosis and facilitate plaque formation and disease progression. Almost all immune system cells, including neutrophils, B cells, T cells monocytes, macrophages, foam cells, and dendritic cells (DCs), play a vital role in atherosclerotic plaque. Atherogenesis, the normal function of the endothelium, is initially disrupted and, then, cells of the immune system are recruited to the endothelium following increased expression of cell adhesion molecules. Accumulation of immune cells and lipids leads to the formation of a necrotic nucleus. As the disease progresses, smooth muscle cells form fibrous layers, whose rupture results in exposing the necrotic nucleus and thrombosis. Accordingly, the present review was conducted to determine the role of different cells in innate and adaptive immune systems in inhibition and progression of atherosclerosis.
Collapse
Affiliation(s)
- Mina Mohmmad‐Rezaei
- Cellular and Molecular Research Center, Basic Health Sciences Institute Shahrekord University of Medical Sciences Shahrekord Iran
| | - Reza Arefnezhad
- Halal Research Center of IRI, FDA Tehran Iran
- Department of Anatomy, School of Medicine Shiraz University of Medical Sciences Shiraz Iran
| | - Reza Ahmadi
- Clinical Biochemistry Research Center, Basic Health Sciences Institute Shahrekord University of Medical Sciences Shahrekord Iran
| | | | - Yousef Mirzaei
- Department of Biogeosciences, Scientific Research Center Soran University Soran Iraq
| | - Mohammad‐Hassan Arjmand
- Cellular and Molecular Research Center, Basic Health Sciences Institute Shahrekord University of Medical Sciences Shahrekord Iran
- Cancer Research Center Shahrekord University of Medical Sciences Shahrekord Iran
| | - Gordon A. Ferns
- Brighton & Sussex Medical School, Division of Medical Education Sussex United Kingdom
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Nader Bagheri
- Cellular and Molecular Research Center, Basic Health Sciences Institute Shahrekord University of Medical Sciences Shahrekord Iran
| |
Collapse
|
6
|
Kandhare AD, Bandyopadhyay D, Thakurdesai PA. Low molecular weight galactomannans-based standardized fenugreek seed extract ameliorates high-fat diet-induced obesity in mice via modulation of FASn, IL-6, leptin, and TRIP-Br2. RSC Adv 2018; 8:32401-32416. [PMID: 35547667 PMCID: PMC9086199 DOI: 10.1039/c8ra05204b] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 09/04/2018] [Indexed: 12/11/2022] Open
Abstract
Background: Obesity is a complex, chronic metabolic disorder and its prevalence is increasing throughout most of the world. Low molecular weight galactomannans-based standardized fenugreek seed extract (LMWGAL-TF) has previously shown anti-diabetic and anti-hyperlipidemic potential. Aim: To evaluate the efficacy and mechanism of action of LMWGAL-TF in treating high fat diet (HFD)-induced obesity and hyperlipidemia in mice. Materials and methods: Male C57BL/6 mice were fed the HFD for 12 weeks and were co-administered with LMWGAL-TF (10, 30 and 100 mg kg-1, p.o.). Variables measured were behavioral, biochemical, molecular and histopathological. In a separate in vitro experiment, copper-ascorbate (Cu-As)-induced mitochondrial oxidative damage was evaluated. Results: The HFD-induced increase (p < 0.001) in body weight, fat mass, lean mass, adipose tissue (brown, mesenteric, epididymal and retroperitoneal) and liver weight was significantly attenuated (p < 0.001) by LMWGAL-TF (30 and 100 mg kg-1). The HFD-induced elevated levels of serum lipid, interleukins (ILs)-6 and leptin were significantly decreased (p < 0.001) by LMWGAL-TF (30 and 100 mg kg-1). Elevated fatty acid synthase (FASn), IL-6, leptin and transcriptional regulator interacting with the PHD-bromodomain 2 (TRIP-Br2) mRNA expression in brown adipose tissue (BAT), liver, and epididymal fat were significantly down-regulated (p < 0.001) by LMWGAL-TF (30 and 100 mg kg-1). Additionally, HFD-induced histological alterations in skeletal muscle, liver, white adipose tissue (WAT) and BAT were also reduced by LMWGAL-TF. Furthermore, the Cu-As-induced alteration in mitochondria oxidative stress (lipid peroxidation, protein carbonylation, glutathione, glutathione reductase, glutathione peroxidase, isocitrate dehydrogenase and α-ketoglutarate dehydrogenase) in skeletal muscle and BAT was significantly (p < 0.001) ameliorated by LMWGAL-TF (2, 4 and 6 mg mL-1) treatment. It also reduced the Cu-As-induced mitochondrial swelling. Conclusion: LMWGAL-TF showed its beneficial effect in reducing HFD-induced obesity via down-regulation of FASn, IL-6, leptin, and TRIP-Br2 in mice.
Collapse
Affiliation(s)
- Amit D Kandhare
- Department of Scientific Affairs, Indus Biotech Private Limited 1, Rahul Residency, Off Salunke Vihar Road, Kondhwa Pune 411048 Maharashtra India +91-9226164041
| | - Debasish Bandyopadhyay
- Oxidative Stress and Free Radical Biology Laboratory, Department of Physiology, University of Calcutta, University College of Science and Technology Kolkata 700 009 India
| | - Prasad A Thakurdesai
- Department of Scientific Affairs, Indus Biotech Private Limited 1, Rahul Residency, Off Salunke Vihar Road, Kondhwa Pune 411048 Maharashtra India +91-9226164041
| |
Collapse
|
7
|
Molica F, Meens MJ, Dubrot J, Ehrlich A, Roth CL, Morel S, Pelli G, Vinet L, Braunersreuther V, Ratib O, Chanson M, Hugues S, Scemes E, Kwak BR. Pannexin1 links lymphatic function to lipid metabolism and atherosclerosis. Sci Rep 2017; 7:13706. [PMID: 29057961 PMCID: PMC5651868 DOI: 10.1038/s41598-017-14130-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 10/06/2017] [Indexed: 12/29/2022] Open
Abstract
Extracellular ATP is a central signaling molecule in inflammatory responses. Pannexin1 (Panx1) channels release ATP in a controlled manner and have been implicated in various inflammatory pathologies, but their role in atherogenesis remains elusive. Using atherosclerosis-susceptible mouse models with ubiquitous deletion of Panx1 (Panx1−/−Apoe−/−) or with Cre recombinase-mediated deletion of Panx1 in endothelial cells and monocytes (Tie2-CreTgPanx1fl/flApoe−/−; Panx1delApoe−/−), we identified a novel role for Panx1 in the lymphatic vasculature. Atherosclerotic lesion development in response to high-cholesterol diet was enhanced in Panx1delApoe−/− mice, pointing to an atheroprotective role for Panx1 in endothelial and/or monocytic cells. Unexpectedly, atherogenesis was not changed in mice with ubiquitous Panx1 deletion, but Panx1−/−Apoe−/− mice displayed reduced body weight, serum cholesterol, triglycerides and free fatty acids, suggesting altered lipid metabolism in these Panx1-deficient mice. Mechanistically, Panx1−/−Apoe−/− mice showed impairment of lymphatic vessel function with decreased drainage of interstitial fluids and reduced dietary fat absorption. Thus, the detrimental effect of Panx1 deletion in endothelial and/or monocytic cells during atherogenesis is counterbalanced by an opposite effect resulting from impaired lymphatic function in ubiquitous Panx1-deficient mice. Collectively, our findings unveil a pivotal role of Panx1 in linking lymphatic function to lipid metabolism and atherosclerotic plaque development.
Collapse
Affiliation(s)
- Filippo Molica
- University of Geneva, Department of Pathology and Immunology, Geneva, CH-1211, Switzerland
| | - Merlijn J Meens
- University of Geneva, Department of Pathology and Immunology, Geneva, CH-1211, Switzerland
| | - Juan Dubrot
- University of Geneva, Department of Pathology and Immunology, Geneva, CH-1211, Switzerland
| | - Avigail Ehrlich
- University of Geneva, Department of Pathology and Immunology, Geneva, CH-1211, Switzerland
| | - Christel L Roth
- University of Geneva, Department of Pathology and Immunology, Geneva, CH-1211, Switzerland
| | - Sandrine Morel
- University of Geneva, Department of Pathology and Immunology, Geneva, CH-1211, Switzerland
| | - Graziano Pelli
- University of Geneva, Department of Pathology and Immunology, Geneva, CH-1211, Switzerland
| | - Laurent Vinet
- Geneva University Hospitals, Department of Radiology and Medical Informatics, Geneva, CH-1211, Switzerland.,University of Geneva and Lausanne, School of Pharmaceutical Sciences, Geneva, CH-1211, Switzerland
| | | | - Osman Ratib
- Geneva University Hospitals, Department of Radiology and Medical Informatics, Geneva, CH-1211, Switzerland
| | - Marc Chanson
- Geneva University Hospitals and University of Geneva, Department of Pediatrics and of Cell Physiology and Metabolism, Geneva, CH-1211, Switzerland
| | - Stephanie Hugues
- University of Geneva, Department of Pathology and Immunology, Geneva, CH-1211, Switzerland
| | - Eliana Scemes
- Albert Einstein College of Medicine, Department of Neuroscience, New York, NY, 10461, USA
| | - Brenda R Kwak
- University of Geneva, Department of Pathology and Immunology, Geneva, CH-1211, Switzerland. .,University of Geneva, Department of Medical Specializations - Cardiology, Geneva, CH-1211, Switzerland.
| |
Collapse
|
8
|
Yan W, Li D, Zhou X. Pravastatin attenuates the action of the ETS domain-containing protein ELK1 to prevent atherosclerosis in apolipoprotein E-knockout mice via modulation of extracellular signal-regulated kinase 1/2 signal pathway. Clin Exp Pharmacol Physiol 2017; 44:344-352. [PMID: 27998006 DOI: 10.1111/1440-1681.12710] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 11/13/2016] [Accepted: 12/12/2016] [Indexed: 12/11/2022]
Abstract
Oxidative stress plays an important role in atherosclerosis, a vascular disease with high morbidity and mortality. The ETS domain-containing protein ELK1 is an oxidative stress-sensitive factor modulated by the extracellular signal-regulated kinase (ERK) 1/2 pathway. However, the role of ELK1 in the prevention of atherosclerosis by pravastatin remains unclear. In the present study, male apolipoprotein E-knockout (apoE-/- ) mice fed a diet containing 1.25% cholesterol (w/w) were divided into two groups, one treated with pravastatin (80 mg/kg, 2-2.4 mg/mouse per day) for 8 weeks and the other not. Male C57BL/6J mice fed with a normal diet were used as a control group. Human umbilical vein endothelial cells (HUVEC) were cultured and treated with pravastatin (10 μmol/L) for 18 hours before testing for the presence or absence of 100 μmol/L H2 O2 (24 hours). Examination of pathological sections from mice aortas revealed that pravastatin treatment almost prevented atherosclerotic plaque formation. Pravastatin also inhibited increases in serum and aortic levels of oxidized low-density lipoprotein and aortic malondialdehyde levels and decreases in aortic reduced glutathione, and the activities of superoxide dismutase, catalase and glutathione peroxidase. H2 O2 -induced increases in reactive oxygen species in HUVECs were reversed by pravastatin by 48%. Pravastatin blocked the phosphorylation of ELK1 and ERK1/2 proteins and reduced mRNA levels of early growth response 1, a known atherogenic transcription factor upregulated by the ROS/ERK/ELK1 pathway, in mice. In conclusion, pravastatin attenuates the action of ELK1 induced by oxidative stress to prevent atherosclerosis, which is dependent partly on modulation of ERK1/2 signalling.
Collapse
Affiliation(s)
- Wei Yan
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dan Li
- Department of Medicine, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Xiaoxu Zhou
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
9
|
Sfyri P, Matsakas A. Crossroads between peripheral atherosclerosis, western-type diet and skeletal muscle pathophysiology: emphasis on apolipoprotein E deficiency and peripheral arterial disease. J Biomed Sci 2017; 24:42. [PMID: 28688452 PMCID: PMC5502081 DOI: 10.1186/s12929-017-0346-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 06/07/2017] [Indexed: 12/16/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory process that, in the presence of hyperlipidaemia, promotes the formation of atheromatous plaques in large vessels of the cardiovascular system. It also affects peripheral arteries with major implications for a number of other non-vascular tissues such as the skeletal muscle, the liver and the kidney. The aim of this review is to critically discuss and assimilate current knowledge on the impact of peripheral atherosclerosis and its implications on skeletal muscle homeostasis. Accumulating data suggests that manifestations of peripheral atherosclerosis in skeletal muscle originates in a combination of increased i)-oxidative stress, ii)-inflammation, iii)-mitochondrial deficits, iv)-altered myofibre morphology and fibrosis, v)-chronic ischemia followed by impaired oxygen supply, vi)-reduced capillary density, vii)- proteolysis and viii)-apoptosis. These structural, biochemical and pathophysiological alterations impact on skeletal muscle metabolic and physiologic homeostasis and its capacity to generate force, which further affects the individual's quality of life. Particular emphasis is given on two major areas representing basic and applied science respectively: a)-the abundant evidence from a well-recognised atherogenic model; the Apolipoprotein E deficient mouse and the role of a western-type diet and b)-on skeletal myopathy and oxidative stress-induced myofibre damage from human studies on peripheral arterial disease. A significant source of reactive oxygen species production and oxidative stress in cardiovascular disease is the family of NADPH oxidases that contribute to several pathologies. Finally, strategies targeting NADPH oxidases in skeletal muscle in an attempt to attenuate cellular oxidative stress are highlighted, providing a better understanding of the crossroads between peripheral atherosclerosis and skeletal muscle pathophysiology.
Collapse
Affiliation(s)
- Peggy Sfyri
- Molecular Physiology Laboratory, Centre for Atherothrombotic & Metabolic Disease, Hull York Medical School, University of Hull, Cottingham Road, Hull, HU6 7RX, United Kingdom
| | - Antonios Matsakas
- Molecular Physiology Laboratory, Centre for Atherothrombotic & Metabolic Disease, Hull York Medical School, University of Hull, Cottingham Road, Hull, HU6 7RX, United Kingdom.
| |
Collapse
|
10
|
Luehmann HP, Detering L, Fors BP, Pressly ED, Woodard PK, Randolph GJ, Gropler RJ, Hawker CJ, Liu Y. PET/CT Imaging of Chemokine Receptors in Inflammatory Atherosclerosis Using Targeted Nanoparticles. J Nucl Med 2016; 57:1124-9. [PMID: 26795285 PMCID: PMC5088780 DOI: 10.2967/jnumed.115.166751] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 12/07/2015] [Indexed: 12/17/2022] Open
Abstract
UNLABELLED Atherosclerosis is inherently an inflammatory process that is strongly affected by the chemokine-chemokine receptor axes regulating the trafficking of inflammatory cells at all stages of the disease. Of the chemokine receptor family, some specifically upregulated on macrophages play a critical role in plaque development and may have the potential to track plaque progression. However, the diagnostic potential of these chemokine receptors has not been fully realized. On the basis of our previous work using a broad-spectrum peptide antagonist imaging 8 chemokine receptors together, the purpose of this study was to develop a targeted nanoparticle for sensitive and specific detection of these chemokine receptors in both a mouse vascular injury model and a spontaneously developed mouse atherosclerosis model. METHODS The viral macrophage inflammatory protein-II (vMIP-II) was conjugated to a biocompatible poly(methyl methacrylate)-core/polyethylene glycol-shell amphiphilic comblike nanoparticle through controlled conjugation and polymerization before radiolabeling with (64)Cu for PET imaging in an apolipoprotein E-deficient (ApoE(-/-)) mouse vascular injury model and a spontaneous ApoE(-/-) mouse atherosclerosis model. Histology, immunohistochemistry, and real-time reverse transcription polymerase chain reaction were performed to assess the plaque progression and upregulation of chemokine receptors. RESULTS The chemokine receptor-targeted (64)Cu-vMIP-II-comb showed extended blood retention and improved biodistribution. PET imaging showed specific tracer accumulation at plaques in ApoE(-/-) mice, confirmed by competitive receptor blocking studies and assessment in wild-type mice. Histopathologic characterization showed the progression of plaque including size and macrophage population, corresponding to the elevated concentration of chemokine receptors and more importantly increased PET signals. CONCLUSION This work provides a useful nanoplatform for sensitive and specific detection of chemokine receptors to assess plaque progression in mouse atherosclerosis models.
Collapse
Affiliation(s)
- Hannah P. Luehmann
- Department of Radiology, Washington University, Campus Box 8225, 510
S. Kingshighway Blvd., St. Louis, MO 63110
| | - Lisa Detering
- Department of Radiology, Washington University, Campus Box 8225, 510
S. Kingshighway Blvd., St. Louis, MO 63110
| | - Brett P. Fors
- Department of Materials, Chemistry and Biochemistry, University of
California, Santa Barbara, California
| | - Eric D. Pressly
- Department of Materials, Chemistry and Biochemistry, University of
California, Santa Barbara, California
| | - Pamela K. Woodard
- Department of Radiology, Washington University, Campus Box 8225, 510
S. Kingshighway Blvd., St. Louis, MO 63110
| | | | - Robert J. Gropler
- Department of Radiology, Washington University, Campus Box 8225, 510
S. Kingshighway Blvd., St. Louis, MO 63110
| | - Craig J. Hawker
- Department of Materials, Chemistry and Biochemistry, University of
California, Santa Barbara, California
| | - Yongjian Liu
- Department of Radiology, Washington University, Campus Box 8225, 510
S. Kingshighway Blvd., St. Louis, MO 63110
| |
Collapse
|
11
|
Cortes R, Ivorra C, Martínez-Hervás S, Pedro T, González-Albert V, Artero A, Adam V, García-García AB, Ascaso JF, Real JT, Chaves FJ. Postprandial Changes in Chemokines Related to Early Atherosclerotic Processes in Familial Hypercholesterolemic Subjects: A Preliminary Study. Arch Med Res 2016; 47:33-9. [DOI: 10.1016/j.arcmed.2016.01.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 01/13/2016] [Indexed: 12/01/2022]
|
12
|
Eikendal ALM, Evelein AMV, Uiterwaal CSPM, van der Ent CK, Visseren FLJ, Bots ML, Hoefer IE, den Ruijter HM, Dalmeijer GW. Relation Between Circulating Inflammatory Chemokines and Vascular Characteristics in Healthy, Young Children. J Am Heart Assoc 2015; 4:e002346. [PMID: 26675251 PMCID: PMC4845277 DOI: 10.1161/jaha.115.002346] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 10/07/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND Atherosclerosis begins in childhood with the occurrence of inflammatory vascular wall alterations that are detectable with B-mode ultrasound. Chemokines appear to be involved in the development of these alterations given that they occur early in the atherosclerotic pathway as mediators of vascular inflammation. However, this has not extensively been investigated. Therefore, we studied in healthy young children whether chemokines monocyte chemotactic protein 1 (MCP-1), regulated on activation normal T-cell expressed and secreted (RANTES), and vascular and intercellular adhesion molecules (VCAM and ICAM) related to vascular characteristics of the carotid artery. METHODS AND RESULTS We obtained demography, anthropometry, and overnight fasting plasma of 139 eight-year-old children of the Wheezing Illnesses Study Leidsche Rijn birth cohort. Carotid intima-media thickness (CIMT), distensibility, and Young's Elastic Modulus (YEM) of the common carotid artery were measured sonographically. Chemokine plasma levels were assessed using a multiplex assay. We studied the relation between the chemokines and vascular characteristics using multivariable linear regression analyses with adjustments for sex, systolic blood pressure, homeostasis model assessment of insulin resistance, triglycerides, low-density lipoprotein- and high-density lipoprotein-cholesterol. Of the studied chemokines, RANTES related to common carotid distensibility and YEM. One standard deviation increase in RANTES level related to a 5.45-MPA(-1) (95% confidence interval [CI], -9.43, -1.39; P=0.01) decrease in distensibility and to a 5.55-kPa increase in YEM (95% CI, 0.40, 10.85; P=0.03). RANTES did not relate to CIMT. MCP-1, VCAM, and ICAM did not relate to any of the studied vascular characteristics. CONCLUSION RANTES appears to be involved in the development of preatherosclerotic inflammatory vascular alterations already in healthy, young children. This may provide further insight into the early-life origins of atherosclerosis.
Collapse
Affiliation(s)
| | - Annemieke M. V. Evelein
- Department of PediatricsWilhelmina Children's HospitalUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Cuno S. P. M. Uiterwaal
- Julius Center for Health Sciences and Primary CareUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Cornelis K. van der Ent
- Department of PediatricsWilhelmina Children's HospitalUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Frank L. J. Visseren
- Department of Vascular MedicineUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Michiel L. Bots
- Julius Center for Health Sciences and Primary CareUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Imo E. Hoefer
- Laboratory of Experimental CardiologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Hester M. den Ruijter
- Laboratory of Experimental CardiologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Geertje W. Dalmeijer
- Julius Center for Health Sciences and Primary CareUniversity Medical Center UtrechtUtrechtThe Netherlands
| |
Collapse
|
13
|
Jeon US, Choi JP, Kim YS, Ryu SH, Kim YK. The enhanced expression of IL-17-secreting T cells during the early progression of atherosclerosis in ApoE-deficient mice fed on a western-type diet. Exp Mol Med 2015; 47:e163. [PMID: 25976521 PMCID: PMC4454994 DOI: 10.1038/emm.2015.19] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 12/02/2014] [Accepted: 01/07/2015] [Indexed: 01/03/2023] Open
Abstract
Atherosclerosis is a chronic progressive inflammatory disorder and the leading cause of cardiovascular mortality. Here we assessed the dynamic changes of T-cell-derived cytokines, such as inteferon (IFN)-γ, interleukin (IL)-17 and IL-4, during the progression of atherosclerosis in apolipoprotein E-null (ApoE−/−) mice, to understand the role of immune responses in different stages of atherosclerosis. Male ApoE−/− mice were fed a high-fat, western-type diet (WD: 21% lipid, 1.5% cholesterol) after 5 weeks of age and were compared with C57BL/6 wild-type control mice fed a standard chow diet. Atherosclerotic lesions appeared in the aortic sinus of ApoE−/− mice 4 weeks after WD and the lesions progressed and occupied >50% of the total sinus area 16 weeks after WD. Aortic IL-17 mRNA and protein expression started to increase in ApoE−/− mice after 4 weeks on the WD and peaked at around 8–12 weeks on the WD. In terms of systemic expression of T-cell-derived cytokines, IL-17 production from splenocytes after anti-CD3/CD28 stimuli increased from 4 weeks on the WD, peaked at 12 weeks and returned to control levels at 16 weeks. The production of IFN-γ and IL-4 (Th1 and Th2 cytokines, respectively) from splenocytes was delayed compared with IL-17. Taken together, the present data indicate that Th17 cell response may be involved at an early stage in the development of atherosclerosis.
Collapse
Affiliation(s)
- Un Sil Jeon
- Division of Molecular and Life Science, Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Jun-Pyo Choi
- Institute of Convergence Medicine, Ewha Womans University School of Medicine, Seoul, Republic of Korea
| | - You-Sun Kim
- Department of Internal Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Sung-Ho Ryu
- Division of Molecular and Life Science, Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Yoon-Keun Kim
- Institute of Convergence Medicine, Ewha Womans University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
14
|
Kotfis K, Biernawska J, Zegan-Barańska M, Żukowski M. Peripheral Blood Lymphocyte Subsets (CD4+, CD8+ T Cells, NK Cells) in Patients with Cardiovascular and Neurological Complications after Carotid Endarterectomy. Int J Mol Sci 2015; 16:10077-94. [PMID: 25946343 PMCID: PMC4463633 DOI: 10.3390/ijms160510077] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 04/20/2015] [Accepted: 04/20/2015] [Indexed: 12/21/2022] Open
Abstract
Background: The aim of the study was to evaluate the differences in the circulating immune cells’ subgroups after the atherosclerotic plaque removal in patients presenting with postoperative complications as compared to the patients without complications after carotid endarterectomy (CEA). Methods: Patients with significant carotid atherosclerosis (n = 124, age range: 44 to 87 years) who underwent CEA were enrolled in a prospective study. The immunology study using flow cytometry was performed to determine the percentages of peripheral blood T cells (CD4+, CD8+, Treg—CD4+/CD25+) and NK (natural killer) cells before and after the procedure. The data were expressed as the percentage of total lymphocytes ± the standard error of mean. Results: The mean percentage of lymphocytes (61.54% ± 17.50% vs. 71.82% ± 9.68%, p = 0.030) and CD4 T lymphocytes (T helper, 38.13% ± 13.78% vs. 48.39% ± 10.24%, p = 0.027) was significantly lower six hours after CEA in patients with postoperative 30-day cardiovascular and neurological complications as compared to the group without complications. On the other hand the mean NK level in the group with complications was significantly higher (21.61% ± 9.00% vs. 15.80% ± 9.31%, p = 0.048). Conclusions: The results of this study suggest that after carotid endarterectomy the percentages of circulating immune cells subsets differ in patients with and without postoperative complications.
Collapse
Affiliation(s)
- Katarzyna Kotfis
- Department of Anaesthesia, Intensive Care and Acute Poisoning, Pomeranian Medical University, Teaching Hospital No. 2, 70-111 Szczecin, Poland.
| | - Jowita Biernawska
- Department of Anaesthesia, Intensive Care and Acute Poisoning, Pomeranian Medical University, Teaching Hospital No. 2, 70-111 Szczecin, Poland.
| | - Małgorzata Zegan-Barańska
- Department of Anaesthesia, Intensive Care and Acute Poisoning, Pomeranian Medical University, Teaching Hospital No. 2, 70-111 Szczecin, Poland.
| | - Maciej Żukowski
- Department of Anaesthesia, Intensive Care and Acute Poisoning, Pomeranian Medical University, Teaching Hospital No. 2, 70-111 Szczecin, Poland.
| |
Collapse
|
15
|
Collins C, Tzima E, Patterson C. Inflammation. Atherosclerosis 2015. [DOI: 10.1002/9781118828533.ch4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
16
|
Kim SM, Kim BY, Lee SA, Eo SK, Yun Y, Kim CD, Kim K. 27-Hydroxycholesterol and 7alpha-hydroxycholesterol trigger a sequence of events leading to migration of CCR5-expressing Th1 lymphocytes. Toxicol Appl Pharmacol 2014; 274:462-70. [DOI: 10.1016/j.taap.2013.12.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 12/10/2013] [Accepted: 12/11/2013] [Indexed: 12/13/2022]
|
17
|
Resveratrol prevents suppression of regulatory T-cell production, oxidative stress, and inflammation of mice prone or resistant to high-fat diet–induced obesity. Nutr Res 2013; 33:971-81. [DOI: 10.1016/j.nutres.2013.07.016] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Revised: 06/28/2013] [Accepted: 07/23/2013] [Indexed: 12/31/2022]
|
18
|
Legein B, Temmerman L, Biessen EAL, Lutgens E. Inflammation and immune system interactions in atherosclerosis. Cell Mol Life Sci 2013; 70:3847-69. [PMID: 23430000 PMCID: PMC11113412 DOI: 10.1007/s00018-013-1289-1] [Citation(s) in RCA: 225] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Revised: 01/30/2013] [Accepted: 02/04/2013] [Indexed: 12/15/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of mortality worldwide, accounting for 16.7 million deaths each year. The underlying cause of the majority of CVD is atherosclerosis. In the past, atherosclerosis was considered to be the result of passive lipid accumulation in the vessel wall. Today's picture is far more complex. Atherosclerosis is considered a chronic inflammatory disease that results in the formation of plaques in large and mid-sized arteries. Both cells of the innate and the adaptive immune system play a crucial role in its pathogenesis. By transforming immune cells into pro- and anti-inflammatory chemokine- and cytokine-producing units, and by guiding the interactions between the different immune cells, the immune system decisively influences the propensity of a given plaque to rupture and cause clinical symptoms like myocardial infarction and stroke. In this review, we give an overview on the newest insights in the role of different immune cells and subtypes in atherosclerosis.
Collapse
Affiliation(s)
- Bart Legein
- Experimental Vascular Pathology, Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Lieve Temmerman
- Experimental Vascular Pathology, Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Erik A. L. Biessen
- Experimental Vascular Pathology, Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Esther Lutgens
- Experimental Vascular Biology, Department of Medical Biochemistry, Academic Medical Center (AMC), University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian’s University, Pettenkoferstrasse 8a/9, 80336 Munich, Germany
| |
Collapse
|
19
|
Abstract
Atherosclerosis is the primary cause of acute myocardial infarction and stroke. It is well established that arterial inflammation in response to accumulation and oxidation of lipoproteins in the vascular wall is the major factor responsible for the development of atherosclerosis. During recent years, it has become apparent that this vascular inflammation is modulated by a complex array of autoimmune responses against modified self-antigens in the atherosclerotic plaque and that both protective and pathogenic immune responses become activated as part of the disease process. Studies of hypercholesterolemia-induced immune activation in mouse models of atherosclerosis have demonstrated that Th1 cells contribute to disease progression while regulatory T cells are protective. It has been suggested that antigen presentation of modified self-antigens in the inflammatory environment of atherosclerotic plaques favors generation of antigen-specific Th1 cells over that of regulatory T cells, resulting in a local loss of tolerance. This concept has stimulated the development of plaque-antigen tolerogenic vaccines to dampen plaque inflammation and disease progression. A first generation of atherosclerosis vaccines based on peptides derived from apoB100 and heat shock proteins have demonstrated promising results in animal studies and are now approaching clinical testing.
Collapse
Affiliation(s)
- Jan Nilsson
- Department of Clinical Sciences, Malmö University Hospital, Lund University, SE-205 02 Malmö, Sweden.
| | | | | |
Collapse
|
20
|
Evani SJ, Dallo SF, Murthy AK, Ramasubramanian AK. Shear Stress Enhances Chemokine Secretion from Chlamydia pneumoniae-infected Monocytes. Cell Mol Bioeng 2013; 6:326-334. [PMID: 24505240 DOI: 10.1007/s12195-013-0291-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Chlamydia pneumoniae is a common respiratory pathogen that is considered a highly likely risk factor for atherosclerosis. C. pneumoniae is disseminated from the lung into systemic circulation via infected monocytes and lodges at the atherosclerotic sites. During transit, C. pneumoniae-infected monocytes in circulation are subjected to shear stress due to blood flow. The effect of mechanical stimuli on infected monocytes is largely understudied in the context of C. pneumoniae infection and inflammation. We hypothesized that fluid shear stress alters the inflammatory response of C. pneumoniae-infected monocytes and contributes to immune cell recruitment to the site of tissue damage. Using an in vitro model of blood flow, we determined that a physiological shear stress of 7.5 dyn/cm2 for 1 h on C. pneumoniae-infected monocytes enhances the production of several chemokines, which in turn is correlated with the recruitment of significantly large number of monocytes. Taken together, these results suggest synergistic interaction between mechanical and chemical factors in C. pneumoniae infection and associated inflammation.
Collapse
Affiliation(s)
- Shankar J Evani
- Department of Biomedical Engineering, and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Shatha F Dallo
- Department of Biomedical Engineering, and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Ashlesh K Murthy
- Departments of Pathology and Dental Medicine, Midwestern University, Downers Grove, IL 60515, USA
| | - Anand K Ramasubramanian
- Department of Biomedical Engineering, and South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX 78249, USA
| |
Collapse
|
21
|
Wei Y, Nazari-Jahantigh M, Chan L, Zhu M, Heyll K, Corbalán-Campos J, Hartmann P, Thiemann A, Weber C, Schober A. The microRNA-342-5p fosters inflammatory macrophage activation through an Akt1- and microRNA-155-dependent pathway during atherosclerosis. Circulation 2013; 127:1609-19. [PMID: 23513069 DOI: 10.1161/circulationaha.112.000736] [Citation(s) in RCA: 183] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Atherosclerosis is a chronic inflammatory vascular disease driven by the subendothelial accumulation of macrophages. The mechanism regulating the inflammatory response in macrophages during atherogenesis remains unclear. Because microRNAs (miRNAs) play a crucial role in cellular signaling by posttranscriptional regulation of gene expression, we studied the miRNA expression profiles during the progression of atherosclerosis. METHODS AND RESULTS Using an miRNA real-time polymerase chain reaction array, we found that macrophage-derived miR-342-5p and miR-155 are selectively upregulated in early atherosclerotic lesions in Apoe(-/-) mice. miR-342-5p directly targets Akt1 through its 3'-untranslated region. Akt1 suppression by miR-342-5p induces proinflammatory mediators such as Nos2 and II6 in macrophages via the upregulation of miR-155. The local application of an miR-342-5p antagomir inhibits the development of atherosclerosis in partially ligated carotid arteries. In atherosclerotic lesions, the miR-342-5p antagomir upregulated Akt1 expression and suppressed the expression of miR-155 and Nos2. This reduced Nos2 expression was associated with a diminished generation of nitrotyrosine in the plaques. Furthermore, systemic treatment with an inhibitor of miR-342-5p reduced the progression of atherosclerosis in the aorta of Apoe(-/-) mice. CONCLUSIONS Macrophage-derived miR-342-5p promotes atherosclerosis and enhances the inflammatory stimulation of macrophages by suppressing the Akt1-mediated inhibition of miR-155 expression. Therefore, targeting miR-342-5p may offer a promising strategy to treat atherosclerotic vascular disease.
Collapse
Affiliation(s)
- Yuanyuan Wei
- Experimental Vascular Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Pettenkoferstrasse 9, 80336 Munich, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Kornete M, Mason ES, Piccirillo CA. Immune Regulation in T1D and T2D: Prospective Role of Foxp3+ Treg Cells in Disease Pathogenesis and Treatment. Front Endocrinol (Lausanne) 2013; 4:76. [PMID: 23805128 PMCID: PMC3691561 DOI: 10.3389/fendo.2013.00076] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 06/08/2013] [Indexed: 12/18/2022] Open
Abstract
There is increasing evidence that dysregulated immune responses play key roles in the pathogenesis and complications of type 1 but also type 2 diabetes. Indeed, chronic inflammation and autoimmunity, which are salient features of type 1 diabetes, are now believed to actively contribute to the pathogenesis of type 2 diabetes. The accumulation of activated innate and adaptive immune cells in various metabolic tissues results in the release of inflammatory mediators, which promote insulin resistance and β-cell damage. Moreover, these dysregulated immune responses can also mutually influence the prevalence of both type 1 and 2 diabetes. In this review article, we discuss the central role of immune responses in the patho-physiology and complications of type 1 and 2 diabetes, and provide evidence that regulation of these responses, particularly through the action of regulatory T cells, may be a possible therapeutic avenue for the treatment of these disease and their respective complications.
Collapse
Affiliation(s)
- Mara Kornete
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
- FOCIS Center of Excellence, Research Institute of the McGill University Health Center, Montreal, QC, Canada
| | - Edward S. Mason
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
- FOCIS Center of Excellence, Research Institute of the McGill University Health Center, Montreal, QC, Canada
| | - Ciriaco A. Piccirillo
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
- FOCIS Center of Excellence, Research Institute of the McGill University Health Center, Montreal, QC, Canada
- *Correspondence: Ciriaco A. Piccirillo, Research Institute of the McGill University Health Center, Montreal General Hospital, 1650 Cedar Avenue, Room L11.132, Montreal, QC H3G 1A4, Canada e-mail:
| |
Collapse
|
23
|
Vasic D, Marx N, Sukhova G, Bach H, Durst R, Grüb M, Hausauer A, Hombach V, Rottbauer W, Walcher D. C-peptide promotes lesion development in a mouse model of arteriosclerosis. J Cell Mol Med 2012; 16:927-35. [PMID: 21707916 PMCID: PMC3822861 DOI: 10.1111/j.1582-4934.2011.01365.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Patients with insulin resistance and early type 2 diabetes exhibit an increased propensity to develop a diffuse and extensive pattern of arteriosclerosis. Typically, these patients show elevated serum levels of the proinsulin cleavage product C-peptide and immunohistochemical data from our group revealed C-peptide deposition in early lesions of these individuals. Moreover, in vitro studies suggest that C-peptide could promote atherogenesis. This study examined whether C-peptide promotes vascular inflammation and lesion development in a mouse model of arteriosclerosis. ApoE-deficient mice on a high fat diet were treated with C-peptide or control injections for 12 weeks and the effect on lesion size and plaque composition was analysed. C-peptide treatment significantly increased C-peptide blood levels by 4.8-fold without having an effect on glucose or insulin levels, nor on the lipid profile. In these mice, C-peptide deposition in atherosclerotic plaques was significantly increased compared with controls. Moreover, lesions of C-peptide-treated mice contained significantly more macrophages (1.6 ± 0.3% versus 0.7 ± 0.2% positive area; P < 0.01) and more vascular smooth muscle cells (4.8 ± 0.6% versus 2.4 ± 0.3% positive area; P < 0.01). Finally, lipid deposition measured by Oil-red-O staining in the aortic arch was significantly higher in the C-peptide group compared with controls. Our results demonstrate that elevated C-peptide levels promote inflammatory cell infiltration and lesion development in ApoE-deficient mice without having metabolic effects. These data obtained in a mouse model of arteriosclerosis support the hypothesis that C-peptide may have an active role in atherogenesis in patients with diabetes and insulin resistance.
Collapse
Affiliation(s)
- Dusica Vasic
- Department of Internal Medicine II - Cardiology, University of Ulm, Ulm, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Wigren M, Nilsson J, Kolbus D. Lymphocytes in atherosclerosis. Clin Chim Acta 2012; 413:1562-8. [PMID: 22565046 DOI: 10.1016/j.cca.2012.04.031] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 04/29/2012] [Accepted: 04/29/2012] [Indexed: 02/08/2023]
Abstract
It is well established that atherosclerosis is caused by an inflammatory process in the arterial intima. However, it is only in recent years that it has become clear that this inflammation is modulated by immune responses against plaque antigens. These antigens are primarily believed to be modified self-antigens such as oxidized LDL. The immune system is challenged to determine whether these antigens should be regarded self and tolerated or non-self and eliminated. The latter will result in plaque development while the first will be protective. T cells are key effectors of both types of responses. An activation of regulatory T cells inhibits auto-reactive T effector cells and is anti-inflammatory. In contrast, if Th1 cells become activated in the plaque this is associated with increased inflammation and disease progression. The role of B cells in atherosclerosis remains to be clarified but some species of athero-protective antibodies have been identified. The elucidation of role of immune system in atherosclerosis has revealed new targets for intervention and both vaccines and antibody-based therapies are presently in or due to enter clinical testing.
Collapse
Affiliation(s)
- Maria Wigren
- Department of Clinical Sciences Malmö, Lund University, Sweden
| | | | | |
Collapse
|
25
|
Norata GD, Pulakazhi Venu VK, Callegari E, Paloschi V, Catapano AL. Effect of Tie-2 conditional deletion of BDNF on atherosclerosis in the ApoE null mutant mouse. Biochim Biophys Acta Mol Basis Dis 2012; 1822:927-35. [PMID: 22386878 DOI: 10.1016/j.bbadis.2012.02.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 01/26/2012] [Accepted: 02/14/2012] [Indexed: 12/16/2022]
Abstract
The reduced expression (haplodeficiency) of the main brain derived neurotrophic factor receptor, namely TrkB is associated with reduced atherosclerosis, smooth muscle cells accumulation and collagen content in the lesion. These data support the concept that brain derived neurotrophic factor of vascular origin may contribute to atherosclerosis. However, to date, no experimental approach was possible to investigate this issue due to the lethality of brain derived neurotrophic factor null mice. To overcome these limitations, we generated a mouse model with a conditional deletion of brain derived neurotrophic factor in endothelial cells (Tie-2 Cre recombinase) on an atherosclerotic prone background (apolipoprotein E knock out) and investigated the effect of conditional brain derived neurotrophic factor deficiency on atherosclerosis. Despite brain derived neurotrophic factor reduction in the vascular wall, mice with conditional deletion of brain derived neurotrophic factor did not develop larger atherosclerotic lesion compared to controls. Smooth muscle cell content as well as the distribution of total and fibrillar collagen was similar in the atherosclerotic lesions from mice with brain derived neurotrophic factor conditional deficiency compared to controls. Finally an extended gene expression analysis failed to identify pro-atherogenic gene expression patterns among the animal with brain derived neurotrophic factor deficiency. In spite of the reduced brain derived neurotrophic factor expression, similar atherosclerosis development was observed in the brain derived neurotrophic factor conditional deficient mouse compared to controls. These pieces of evidence indicate that endothelial derived-brain derived neurotrophic factor is not a pro-atherogenic factor and would rather suggest to investigate the role of other TrkB activators on atherosclerosis.
Collapse
|
26
|
Abstract
Cardiovascular disease, a leading cause of mortality worldwide, is caused mainly by atherosclerosis, a chronic inflammatory disease of blood vessels. Lesions of atherosclerosis contain macrophages, T cells and other cells of the immune response, together with cholesterol that infiltrates from the blood. Targeted deletion of genes encoding costimulatory factors and proinflammatory cytokines results in less disease in mouse models, whereas interference with regulatory immunity accelerates it. Innate as well as adaptive immune responses have been identified in atherosclerosis, with components of cholesterol-carrying low-density lipoprotein triggering inflammation, T cell activation and antibody production during the course of disease. Studies are now under way to develop new therapies based on these concepts of the involvement of the immune system in atherosclerosis.
Collapse
|
27
|
Imai T, Oikawa Y, Shimada A, Oguchi S, Takamiya Y, Katsuki T, Okubo Y, Osaki T, Tahara H, Matsushima Y, Miyazaki JI, Itoh H. Proatherogenic Effect of Interleukin-18 is Exerted with High-fat Diet, but not with Normal Diet in Spontaneously Hyperlipidemic Mice. J Atheroscler Thromb 2011; 18:1090-101. [DOI: 10.5551/jat.7567] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
28
|
Brochériou I, Maouche S, Durand H, Braunersreuther V, Le Naour G, Gratchev A, Koskas F, Mach F, Kzhyshkowska J, Ninio E. Antagonistic regulation of macrophage phenotype by M-CSF and GM-CSF: implication in atherosclerosis. Atherosclerosis 2010; 214:316-24. [PMID: 21159337 DOI: 10.1016/j.atherosclerosis.2010.11.023] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Revised: 11/05/2010] [Accepted: 11/21/2010] [Indexed: 10/18/2022]
Abstract
OBJECTIVES We characterized the transcriptional profiles of GM-CSF- (GM-MØ) and M-CSF-induced macrophages (M-MØ) and investigated in situ a subset of differentially expressed genes in human and mouse atherosclerotic lesions. METHODS AND RESULTS Using microarrays we identified a number of genes and biological processes differentially regulated in M-MØ vs GM-MØ. By varying in culture the M-CSF/GM-CSF ratio (0-10), a spectrum of macrophage phenotypes was explored by RT-QPCR. M-CSF (10 ng/ml) stimulated expression of several genes, including selenoprotein-1 (SEPP1), stabilin-1 (STAB1) and CD163 molecule-like-1 (CD163L1) which was inhibited by a low dose of GM-CSF (1 ng/ml); M-CSF inhibited the expression of pro-platelet basic protein (PPBP) induced by GM-CSF. Combining tissue microarrays/quantitative immunohistochemistry of human aortic lesions with RT-QPCR expression data either from human carotids vs mammary non-atherosclerotic arteries or from the apoE null mice normal and atherosclerotic aortas showed that, STAB1, SEPP1 and CD163L1 (M-CSF-sensitive genes) and PPBP (GM-CSF-sensitive gene) were expressed in both human arterial and apoE null mice atherosclerotic tissues. CONCLUSION A balance between M-CSF vs GM-CSF defines macrophage functional polarisation and may contribute to the progression of atherosclerosis.
Collapse
Affiliation(s)
- Isabelle Brochériou
- INSERM UMRS937, Université Pierre et Marie Curie UPMC-Paris 6, Faculté de Médecine Pierre et Marie Curie, 91 Boulevard de l'Hôpital, 75634 Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Inhibition of atherosclerotic lesion development in the ApoE-/- mouse by a novel β-oxa polyunsaturated fatty acid. J Cardiovasc Pharmacol 2010; 56:431-9. [PMID: 20930595 DOI: 10.1097/fjc.0b013e3181f1d420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Recent findings that a novel polyunsaturated fatty acid, β-oxa 23:4n-6, inhibits adhesion molecule expression on vascular endothelial cells and leukocyte adhesion led us to examine its ability to inhibit the development of atherosclerosis in the apoE-deficient (apoE) mouse. The mice were kept on normal chow or a high-fat/high-cholesterol diet for various periods and treated with either vehicle or β-oxa 23:4n-6 by the intraperitoneal route. The hearts and aortae were isolated and lesion development at the aortic root was determined. Morphometric assessment revealed that lesion development was a function of compensatory aortic enlargement, suggesting that measurement of plaque size per se is the appropriate assessment of lesion size. Using this criterion, we found that atherosclerosis development was reduced in response to β-oxa 23:4n-6, plaque size by 74% and aortic cross-sectional area by 62%, under an optimized regime. The number of foam cells per unit tissue area in the lesions of β-oxa 23:4n-6-treated mice was significantly reduced by 37.5%. The blood levels of β-oxa23:4n-6 in these mice exceeded the concentrations previously found to inhibit adhesion molecule expression in cultured endothelial cells. These data show that β-oxa23:4n-6 protects against experimental atherosclerosis, most likely by reducing the number of infiltrating monocytes.
Collapse
|
30
|
Daissormont ITMN, Kraaijeveld AO, Biessen EAL. Chemokines as therapeutic targets for atherosclerotic plaque destabilization and rupture. Future Cardiol 2010; 5:273-84. [PMID: 19450053 DOI: 10.2217/fca.09.4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Chemokines are instrumental in the initiation and progression of atherosclerosis. Recent advances in genomic technologies and the recognition of atherosclerosis as an inflammatory disease have given great impetus to studies addressing the relevance of chemokines for the clinically manifest stages of atherosclerosis and acute cardiovascular syndromes. In this paper, we will review the current status of these studies, highlighting those chemokines that have already been associated with plaque destabilization and rupture. We will recapitulate recent epidemiologic, genomic, histopathological and experimental support for the prominent role of particular chemokines in acute cardiovascular syndromes. Collectively, these data underpin the potential of chemokines as biomarkers and/or therapeutic targets, but also expose the lacunae in our understanding of the precise function of chemokines in the atherosclerosis-related disorders and in the efficacy of chemokine-targeted clinical trials.
Collapse
Affiliation(s)
- Isabelle T M N Daissormont
- Department of Pathology, Maastricht University Medical Center, P Debyelaan 25, Maastricht 6229 HX, The Netherlands.
| | | | | |
Collapse
|
31
|
Inflammatory activation in children with primary hypertension. Pediatr Nephrol 2010; 25:1711-8. [PMID: 20495830 DOI: 10.1007/s00467-010-1548-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2010] [Revised: 03/03/2010] [Accepted: 04/17/2010] [Indexed: 12/14/2022]
Abstract
Low-grade inflammation plays a role in the pathogenesis of primary hypertension (PH) and target organ damage (TOD). We evaluated the profile of inflammatory mediators (CRP, RANTES, MIP-1beta, MIP-1alpha, MCP-1, IL-6, angiogenin, adiponectin) in 30 healthy children (12.7 +/- 3.3 years) and 44 patients with untreated PH (13.7 +/- 2.7 years; n.s). Patients had greater concentrations of CRP, MIP-1beta, and RANTES than controls (all p < 0.05). Children with metabolic syndrome (MS) had greater CRP than children without MS (p = 0.007) and CRP correlated with number of MS criteria, body mass index (BMI), visceral fat, deep subcutaneous fat assessed by magnetic resonance imaging, carotid intima-media thickness (cIMT), left ventricular mass index, and markers of oxidative stress. RANTES correlated with cholesterol, LDL cholesterol, ApoB, and ApoB/ApoA1. Angiogenin correlated with BMI, waist circumference, visceral fat, uric acid, and patients with cIMT>2SD had greater concentration of angiogenin than those with normal cIMT (p = 0.03). Adiponectin was lower in patients with cIMT>2SD than in those with normal cIMT (p = 0.02). No model explaining variability of TOD has been built. Elevated RANTES and MIP-1beta and normal IL-6 and TNF-alpha levels indicate a vascular inflammatory process. Lack of correlation between CRP and chemokines suggests that vascular inflammation in PH precedes the systemic inflammatory changes.
Collapse
|
32
|
Floyd HS, Chen LC, Vallanat B, Dreher K. Fine ambient air particulate matter exposure induces molecular alterations associated with vascular disease progression within plaques of atherosclerotic susceptible mice. Inhal Toxicol 2010; 21:394-403. [PMID: 19496694 DOI: 10.1080/08958370802317745] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Epidemiology studies have reported associations between increased mortality and morbidity with exposure to particulate air pollution, particularly within individuals with preexisting cardiovascular disease (CVD). Clinical and toxicological studies have provided evidence that exposure to ambient air particulate matter (PM) impacts CVD by increasing plaque size. It is unclear whether PM-induced increased plaque size is associated with molecular disease progression. This study examines molecular profiles within plaques recovered from ApoE(-/-) mice exposed to concentrated ambient air particles (CAPs) to determine whether pulmonary deposition of PM contributes to molecular alterations leading to vascular disease progression. Laser capture microdissection was used to recover atherosclerotic plaques from ApoE(-/-) male mice exposed daily for 5 mo to filtered air or CAPs. Alterations in mRNA expression was assessed in microdissected plaques of CAPs-exposed and air controls using the Affymetrix microarray platform. Bioinformatic analysis indicated alterations in 611 genes: 395 genes downregulated and 216 genes upregulated. Gene ontology revealed CAPs-induced changes to inflammation, proliferation, cell cycle, hematological system, and cardiovascular pathways. Quantitative reverse-transcription polymerase chain reaction (qRT-PCR) verified microarray data also revealing gene expression alterations undetected by the microarray analysis, i.e., decreased expression of alpha-actin for smooth muscle cells, and increased expression of the macrophage marker Cd68 and of beta-actin. Comparison of CAPs-induced gene expression profiles demonstrated consistency with previously published gene expression profiles in the ApoE(-/-) mouse model and humans associated with plaque progression. These results indicate that exposure to fine PM induces molecular alterations associated with vascular disease progression and provides insight into potential biological pathways responsible for this effect.
Collapse
Affiliation(s)
- Heather S Floyd
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | | | | | | |
Collapse
|
33
|
Tibolla G, Norata G, Meda C, Arnaboldi L, Uboldi P, Piazza F, Ferrarese C, Corsini A, Maggi A, Vegeto E, Catapano A. Increased atherosclerosis and vascular inflammation in APP transgenic mice with apolipoprotein E deficiency. Atherosclerosis 2010; 210:78-87. [DOI: 10.1016/j.atherosclerosis.2009.10.040] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2009] [Revised: 10/29/2009] [Accepted: 10/30/2009] [Indexed: 11/30/2022]
|
34
|
Braunersreuther V, Pellieux C, Pelli G, Burger F, Steffens S, Montessuit C, Weber C, Proudfoot A, Mach F, Arnaud C. Chemokine CCL5/RANTES inhibition reduces myocardial reperfusion injury in atherosclerotic mice. J Mol Cell Cardiol 2010; 48:789-98. [DOI: 10.1016/j.yjmcc.2009.07.029] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Accepted: 07/29/2009] [Indexed: 10/20/2022]
|
35
|
Chadjichristos CE, Scheckenbach KEL, van Veen TAB, Richani Sarieddine MZ, de Wit C, Yang Z, Roth I, Bacchetta M, Viswambharan H, Foglia B, Dudez T, van Kempen MJA, Coenjaerts FEJ, Miquerol L, Deutsch U, Jongsma HJ, Chanson M, Kwak BR. Endothelial-specific deletion of connexin40 promotes atherosclerosis by increasing CD73-dependent leukocyte adhesion. Circulation 2010; 121:123-31. [PMID: 20026782 DOI: 10.1161/circulationaha.109.867176] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Endothelial dysfunction is the initiating event of atherosclerosis. The expression of connexin40 (Cx40), an endothelial gap junction protein, is decreased during atherogenesis. In the present report, we sought to determine whether Cx40 contributes to the development of the disease. METHODS AND RESULTS Mice with ubiquitous deletion of Cx40 are hypertensive, a risk factor for atherosclerosis. Consequently, we generated atherosclerosis-susceptible mice with endothelial-specific deletion of Cx40 (Cx40del mice). Cx40del mice were indeed not hypertensive. The progression of atherosclerosis was increased in Cx40del mice after 5 and 10 weeks of a high-cholesterol diet, and spontaneous lesions were observed in the aortic sinuses of young mice without such a diet. These lesions showed monocyte infiltration into the intima, increased expression of vascular cell adhesion molecule-1, and decreased expression of the ecto-enzyme CD73 in the endothelium. The proinflammatory phenotype of Cx40del mice was confirmed in another model of induced leukocyte recruitment from the lung microcirculation. Endothelial CD73 is known to induce antiadhesion signaling via the production of adenosine. We found that reducing Cx40 expression in vitro with small interfering RNA or antisense decreased CD73 expression and activity and increased leukocyte adhesion to mouse endothelial cells. These effects were reversed by an adenosine receptor agonist. CONCLUSIONS Cx40-mediated gap junctional communication contributes to a quiescent nonactivated endothelium by propagating adenosine-evoked antiinflammatory signals between endothelial cells. Alteration in this mechanism by targeting Cx40 promotes leukocyte adhesion to the endothelium, thus accelerating atherosclerosis.
Collapse
Affiliation(s)
- C E Chadjichristos
- Division of Cardiology, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Viel EC, Lemarié CA, Benkirane K, Paradis P, Schiffrin EL. Immune regulation and vascular inflammation in genetic hypertension. Am J Physiol Heart Circ Physiol 2009; 298:H938-44. [PMID: 20044442 DOI: 10.1152/ajpheart.00707.2009] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Immune cells have been implicated in the pathogenesis of hypertension. We hypothesized that under the influence of chromosome (chr)2, T lymphocytes contribute to vascular inflammation in genetic salt-sensitive hypertension. Normotensive (Brown Norway), hypertensive (Dahl salt-sensitive), and consomic rats (SSBN2; in which chr2 has been transferred from Brown Norway to Dahl rats) were studied. Systolic blood pressure, measured by tail cuff, and aortic preproendothelin mRNA, measured by quantitative RT-PCR, were elevated in Dahl rats compared with Brown Norway rats and were reduced in SSBN2 rats compared with Dahl rats (P < 0.01). Compared with Brown Norway rats, Dahl rats exhibited increased inflammatory markers and mediators such as nuclear translocation of the aortic p65 subunit of NF-kappaB as well as VCAM-1, ICAM-1, chemokine (C-C motif) receptor 5, and CD4 mRNA, all of which were reduced in SSBN2 rats. Aortic CD8 mRNA was equally increased in Dahl and SSBN2 rats relative to Brown Norway rats. CD4(+) T cell infiltration in the aorta of SSBN2 rats was reduced compared with Dahl rats, whereas the aortic protein expression of Foxp3b and immunosuppressors transforming growth factor (TGF)-beta(1) and IL-10, the three markers associated with the regulatory T cell lineage, were enhanced in SSBN2 rats. Activation in vitro of T cells demonstrated that CD4(+)CD25(+) and CD8(+)CD25(+) cells (Tregs) produce IL-10 in SSBN2 rats. Thus, increased vascular inflammatory responses and hypertension in a genetic salt-sensitive hypertensive rodent model are reduced by transfer of chr2 from a normotensive strain, and this is associated with enhanced levels of immunosuppressive mediators.
Collapse
Affiliation(s)
- Emilie C Viel
- Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | | | | | | | | |
Collapse
|
37
|
Andersson J, Libby P, Hansson GK. Adaptive immunity and atherosclerosis. Clin Immunol 2009; 134:33-46. [PMID: 19635683 DOI: 10.1016/j.clim.2009.07.002] [Citation(s) in RCA: 211] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2009] [Revised: 06/30/2009] [Accepted: 07/01/2009] [Indexed: 12/12/2022]
Abstract
Atherosclerosis involves the formation of inflammatory arterial lesions and is one of the most common causes of death globally. It has been evident for more than 20 years that adaptive immunity and T cells in particular regulate the magnitude of the atherogenic pro-inflammatory response. T cells also influence the stability of the atherosclerotic lesion and thus the propensity for thrombus formation and the clinical outcome of disease. This review summarizes our current understanding of T cells in atherogenesis, including which antigens they recognize, the role of T cell costimulation/coinhibition, and their secretion of pro- and anti-inflammatory mediators. Furthermore, we outline future areas of research and potential clinical intervention strategies.
Collapse
Affiliation(s)
- John Andersson
- Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm SE-17176, Sweden.
| | | | | |
Collapse
|
38
|
Abstract
Atherosclerosis, the cause of myocardial infarction, stroke and ischemic gangrene, is an inflammatory disease. When LDL accumulates in the intima, it activates the endothelium to express leukocyte adhesion molecules and chemokines that promote recruitment of monocytes and T cells. Monocyte-derived macrophages upregulate pattern recognition receptors, including scavenger receptors that mediate uptake of modified LDL, and Toll-like receptors, which transmit activating signals leading to release of cytokines, proteases, and vasoactive molecules. T cells in lesions recognize local antigens and mount Th1 responses with secretion of pro-inflammatory cytokines, thus contributing to local inflammation and growth of the plaque. Intensified inflammatory activation may lead to local proteolysis, plaque rupture, and thrombus formation, triggering ischemia and infarction. Inflammatory markers are already used to monitor the disease process and anti-inflammatory therapy may be useful to control disease activity.
Collapse
Affiliation(s)
- G K Hansson
- Karolinska Institutet, Center for Molecular Medicine L8:03, Department of Medicine, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
39
|
Lee J, Zhuang Y, Wei X, Shang F, Wang J, Zhang Y, Liu X, Yang Y, Liu L, Zheng Q. Contributions of PD-1/PD-L1 pathway to interactions of myeloid DCs with T cells in atherosclerosis. J Mol Cell Cardiol 2009; 46:169-76. [DOI: 10.1016/j.yjmcc.2008.10.028] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2008] [Revised: 10/28/2008] [Accepted: 10/31/2008] [Indexed: 12/01/2022]
|
40
|
Zhou XX, Gao PJ, Sun BG. Pravastatin attenuates interferon-gamma action via modulation of STAT1 to prevent aortic atherosclerosis in apolipoprotein E-knockout mice. Clin Exp Pharmacol Physiol 2008; 36:373-9. [PMID: 19018808 DOI: 10.1111/j.1440-1681.2008.05067.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
1. The beneficial effects of pravastatin, beyond that of lowering cholesterol in atherosclerosis, include reducing the action of interferon (IFN)-gamma. Interferon-gamma activates the signal transducer and activator of transcription 1 (STAT1), but it is unclear whether the inhibitory effect of pravastatin in atherosclerosis is via modulation of the IFN-gamma/STAT1 pathway. Thus, the aim of the present study was to determine whether the action of pravastatin in preventing aortic atherosclerosis by attenuation of IFN-gamma action is dependent on STAT1. 2. Male apolipoprotein E-knockout (apoE(-/-)) mice were fed a diet containing 1.25% cholesterol (w/w). Mice were divided into two groups, one of which was supplemented with pravastatin (80 mg/kg per day). Male C57BL/6J mice were fed a normal diet and served as the control group (n = 12 per group). 3. Atherosclerotic lesions in the aortic root were assessed by staining sections haematoxylin and eosin. Serum concentrations of IFN-gamma and IFN-gamma mRNA expression in the thoracoabdominal aorta were determined by ELISA and real-time quantitative polymerase chain reaction methods, respectively. Expression of phosphorylated STAT1 (pSTAT1), interferon regulating factor (IRF)-1 and suppressors of cytokine signalling 1 (SOCS1) was determined in the thoracoabdominal aorta using Western blot analysis. 4. After 8 weeks, pravastatin treatment significantly prevented the formation of atherosclerotic lesions (P < 0.05) and reduced serum IFN-gamma concentrations (P < 0.05) and levels of IFN-gamma mRNA within the aorta (P < 0.01). Pravastatin significantly decreased the expressions of pSTAT1 and IRF-1 within the aorta and significantly increased expression of SOCS1. 5. These results suggest that the actions of pravastatin in attenuating the action of IFN-gamma and subsequently preventing aortic atherosclerosis may depend, at least in part, on modulation of STAT1 activity. This providing us with a new therapeutic approach and a clearer insight into the clinical benefits of pravastatin.
Collapse
Affiliation(s)
- Xiao-Xu Zhou
- Department of Cardiology, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| | | | | |
Collapse
|
41
|
Braunersreuther V, Steffens S, Arnaud C, Pelli G, Burger F, Proudfoot A, Mach F. A novel RANTES antagonist prevents progression of established atherosclerotic lesions in mice. Arterioscler Thromb Vasc Biol 2008; 28:1090-6. [PMID: 18388327 DOI: 10.1161/atvbaha.108.165423] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND Atherosclerosis is a chronic inflammatory disease that represents the primary cause of death through coronary disease and stroke. Chemokines are known to play a crucial role in this disease by recruiting inflammatory leukocytes to the endothelium. Recently, the chemokine variant [44AANA47]-RANTES was shown to impair inflammatory cell recruitment in vivo by interfering with heparin binding and oligomerization. METHODS AND RESULTS In this study we report that curative treatment with [44AANA47]-RANTES limits atherosclerotic plaque formation in LDLr-/- mice. This was associated with reduced infiltration of T cells and macrophages and reduced production of matrix metalloproteinase (MMP)-9. By contrast, the relative smooth muscle cell and collagen content was increased, indicating a more stable plaque phenotype. In addition, we provide evidence for direct inhibition of leukocyte recruitment into aortic root lesions, attenuated leukocyte rolling and arrest in mesenteric vessels, as well as a reduced proinflammatory response following Con A stimulation in vitro. CONCLUSIONS Interference with chemokine oligomerization and chemokine/heparin interactions is a powerful novel approach that inhibits progression of established atherosclerosis in mice. By inhibiting leukocyte recruitment into plaques, [44AANA47]-RANTES mediates a less inflammatory plaque phenotype and thus reduced systemic inflammatory state.
Collapse
Affiliation(s)
- Vincent Braunersreuther
- Division of Cardiology, Department of Medicine, University Hospital, Foundation for Medical Researches, 64 Avenue Roseraie, 1211 Geneva, Switzerland
| | | | | | | | | | | | | |
Collapse
|
42
|
Watari Y, Yamamoto Y, Brydun A, Ishida T, Mito S, Yoshizumi M, Igarashi K, Chayama K, Ohshima T, Ozono R. Ablation of the Bach1 Gene Leads to the Suppression of Atherosclerosis in Bach1 and Apolipoprotein E Double Knockout Mice. Hypertens Res 2008; 31:783-92. [DOI: 10.1291/hypres.31.783] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
43
|
Hansson GK, Robertson AKL, Söderberg-Nauclér C. Inflammation and atherosclerosis. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2007; 1:297-329. [PMID: 18039117 DOI: 10.1146/annurev.pathol.1.110304.100100] [Citation(s) in RCA: 757] [Impact Index Per Article: 42.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Atherosclerosis, the cause of myocardial infarction, stroke, and ischemic gangrene, is an inflammatory disease. The atherosclerotic process is initiated when cholesterol-containing low-density lipoproteins accumulate in the intima and activate the endothelium. Leukocyte adhesion molecules and chemokines promote recruitment of monocytes and T cells. Monocytes differentiate into macrophages and upregulate pattern recognition receptors, including scavenger receptors and toll-like receptors. Scavenger receptors mediate lipoprotein internalization, which leads to foam-cell formation. Toll-like receptors transmit activating signals that lead to the release of cytokines, proteases, and vasoactive molecules. T cells in lesions recognize local antigens and mount T helper-1 responses with secretion of pro-inflammatory cytokines that contribute to local inflammation and growth of the plaque. Intensified inflammatory activation may lead to local proteolysis, plaque rupture, and thrombus formation, which causes ischemia and infarction. Inflammatory markers are already used to monitor the disease process and anti-inflammatory therapy may be useful to control disease activity.
Collapse
Affiliation(s)
- Göran K Hansson
- Department of Medicine, Center for Molecular Medicine, Karolinska Institute, Stockholm SE-17176, Sweden.
| | | | | |
Collapse
|
44
|
Nelson WD, Zenovich AG, Ott HC, Stolen C, Caron GJ, Panoskaltsis-Mortari A, Barnes SA, Xin X, Taylor DA. Sex-Dependent Attenuation of Plaque Growth After Treatment With Bone Marrow Mononuclear Cells. Circ Res 2007; 101:1319-27. [DOI: 10.1161/circresaha.107.155564] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
There are clinically relevant differences in symptomatology, risk stratification, and efficacy of therapies between men and women with coronary artery disease. Sex-based differences in plaque attenuation after administration of bone marrow mononuclear cells (BMNCs) are unknown. Forty-five male and 57 female apolipoprotein-E knockout (apoE
−/−
) mice were fed a high-fat diet. At 14 weeks of age, animals received 4 biweekly intravenous sex-matched (males, n=11; females, n=13) or -mismatched (males, n=12; females, n=14) BMNCs obtained from C57BL6/J mice. The rest of the apoE
−/−
mice were vehicle treated (males, n=13; females, n=20) or were age-matched untreated controls (males, n=9; females, n=10). Aortic plaque burden, progenitor cell profiles in bone marrow (BM) and 22 circulating cytokines/chemokines were examined 1 week following the final injection. Only female BMNCs infused into male apoE
−/−
recipients significantly decreased plaque formation (
P
<0.001). This reparative response univariately correlated with increased CD34
+
(
P
=0.02), CD45
+
(
P
=0.0001), and AC133
+
/CD34
+
(
P
=0.001) cell percentages in the BM of recipients but not with total serum cholesterol or percentage of BM-CD31
+
/CD45
low
cells. In a multivariate analysis, BM-AC133
+
/CD34
+
and BM-CD45
+
percentage counts correlated with a lower plaque burden (
P
<0.05). Increased granulocyte colony-stimulating factor levels highly correlated with plaque attenuation (
r
=−0.86,
P
=0.0004). In untreated apoE
−/−
mice of either sex, BM-AC133
+
/CD34
+
cells rose initially and then fell as plaque accumulated; however, BM-AC133
+
/CD34
+
percentages were higher in females at all times (
P
≤0.01). We have demonstrated an atheroprotective effect of female-derived BMNCs administered to male atherosclerotic apoE
−/−
mice; this reparative response correlated with the upregulation of BM-AC133
+
/CD34
+
and CD45
+
cells and of circulating granulocyte colony-stimulating factor. Atherosclerotic female apoE
−/−
mice did not exhibit atheroprotection after BMNCs of either sex. Our findings may have implications for clinical cell therapy trials for coronary artery disease. Further exploration of sex-based differences in atheroprotection and vascular repair is warranted.
Collapse
Affiliation(s)
- Wendy D. Nelson
- From the Center for Cardiovascular Repair (A.G.Z., G.J.C., D.A.T.), Department of Pediatrics (A.P.M.), and the School of Mathematics and Department of Biomedical Engineering (X.X.), University of Minnesota, Minneapolis; R&D Systems (W.D.N.), Minneapolis, Minn; Massachusetts General Hospital (H.C.O.), Boston; Boston Scientific Corporation (G.S.), Natick, Mass; and the School of Nursing (S.A.B.), University of Arkansas, Fayetteville
| | - Andrey G. Zenovich
- From the Center for Cardiovascular Repair (A.G.Z., G.J.C., D.A.T.), Department of Pediatrics (A.P.M.), and the School of Mathematics and Department of Biomedical Engineering (X.X.), University of Minnesota, Minneapolis; R&D Systems (W.D.N.), Minneapolis, Minn; Massachusetts General Hospital (H.C.O.), Boston; Boston Scientific Corporation (G.S.), Natick, Mass; and the School of Nursing (S.A.B.), University of Arkansas, Fayetteville
| | - Harald C. Ott
- From the Center for Cardiovascular Repair (A.G.Z., G.J.C., D.A.T.), Department of Pediatrics (A.P.M.), and the School of Mathematics and Department of Biomedical Engineering (X.X.), University of Minnesota, Minneapolis; R&D Systems (W.D.N.), Minneapolis, Minn; Massachusetts General Hospital (H.C.O.), Boston; Boston Scientific Corporation (G.S.), Natick, Mass; and the School of Nursing (S.A.B.), University of Arkansas, Fayetteville
| | - Craig Stolen
- From the Center for Cardiovascular Repair (A.G.Z., G.J.C., D.A.T.), Department of Pediatrics (A.P.M.), and the School of Mathematics and Department of Biomedical Engineering (X.X.), University of Minnesota, Minneapolis; R&D Systems (W.D.N.), Minneapolis, Minn; Massachusetts General Hospital (H.C.O.), Boston; Boston Scientific Corporation (G.S.), Natick, Mass; and the School of Nursing (S.A.B.), University of Arkansas, Fayetteville
| | - Gabriel J. Caron
- From the Center for Cardiovascular Repair (A.G.Z., G.J.C., D.A.T.), Department of Pediatrics (A.P.M.), and the School of Mathematics and Department of Biomedical Engineering (X.X.), University of Minnesota, Minneapolis; R&D Systems (W.D.N.), Minneapolis, Minn; Massachusetts General Hospital (H.C.O.), Boston; Boston Scientific Corporation (G.S.), Natick, Mass; and the School of Nursing (S.A.B.), University of Arkansas, Fayetteville
| | - Angela Panoskaltsis-Mortari
- From the Center for Cardiovascular Repair (A.G.Z., G.J.C., D.A.T.), Department of Pediatrics (A.P.M.), and the School of Mathematics and Department of Biomedical Engineering (X.X.), University of Minnesota, Minneapolis; R&D Systems (W.D.N.), Minneapolis, Minn; Massachusetts General Hospital (H.C.O.), Boston; Boston Scientific Corporation (G.S.), Natick, Mass; and the School of Nursing (S.A.B.), University of Arkansas, Fayetteville
| | - Samuel A. Barnes
- From the Center for Cardiovascular Repair (A.G.Z., G.J.C., D.A.T.), Department of Pediatrics (A.P.M.), and the School of Mathematics and Department of Biomedical Engineering (X.X.), University of Minnesota, Minneapolis; R&D Systems (W.D.N.), Minneapolis, Minn; Massachusetts General Hospital (H.C.O.), Boston; Boston Scientific Corporation (G.S.), Natick, Mass; and the School of Nursing (S.A.B.), University of Arkansas, Fayetteville
| | - Xiangrong Xin
- From the Center for Cardiovascular Repair (A.G.Z., G.J.C., D.A.T.), Department of Pediatrics (A.P.M.), and the School of Mathematics and Department of Biomedical Engineering (X.X.), University of Minnesota, Minneapolis; R&D Systems (W.D.N.), Minneapolis, Minn; Massachusetts General Hospital (H.C.O.), Boston; Boston Scientific Corporation (G.S.), Natick, Mass; and the School of Nursing (S.A.B.), University of Arkansas, Fayetteville
| | - Doris A. Taylor
- From the Center for Cardiovascular Repair (A.G.Z., G.J.C., D.A.T.), Department of Pediatrics (A.P.M.), and the School of Mathematics and Department of Biomedical Engineering (X.X.), University of Minnesota, Minneapolis; R&D Systems (W.D.N.), Minneapolis, Minn; Massachusetts General Hospital (H.C.O.), Boston; Boston Scientific Corporation (G.S.), Natick, Mass; and the School of Nursing (S.A.B.), University of Arkansas, Fayetteville
| |
Collapse
|
45
|
Abstract
Atherosclerosis is a chronic inflammatory disease that is the primary cause of myocardial infarction and stroke, which occur after sudden thrombotic occlusion of an artery. A growing body of evidence suggests that cannabinoid signalling plays a fundamental role in atherosclerosis development and its clinical manifestations. Thus, CB2 receptors are protective in myocardial ischaemia/reperfusion and implicated in the modulation of chemotaxis, which is crucial for the recruitment of leukocytes during inflammation. Delta-9-Tetrahydrocannabinol (THC)-mediated activation has been shown to inhibit atherosclerotic plaque progression in a CB2 dependent manner. Although CB1 and CB2 expression has been reported on platelets, their involvement in thrombus formation is still controversial. While several reports suggest that CB1 receptors may have a relevant role in neuroprotection after ischaemic stroke, recent studies show the protective effects in various forms of neuroprotection are not related to CB1 stimulation, and a protective role of CB1 blockade has also been reported. In addition, vascular and myocardial CB1 receptors contribute to the modulation of blood pressure and heart rate. It is tempting to suggest that pharmacological modulation of the endocannabinoid system is a potential novel therapeutic strategy in the treatment of atherosclerosis. For these purposes, it is important to better understand the complex mechanisms of endocannabinoid signalling and potential consequences of its pharmacological modulation, as it may have both pro- and anti-atherosclerotic effects.
Collapse
|
46
|
Joven J, Rull A, Ferré N, Escolà-Gil JC, Marsillach J, Coll B, Alonso-Villaverde C, Aragones G, Claria J, Camps J. The results in rodent models of atherosclerosis are not interchangeable: the influence of diet and strain. Atherosclerosis 2007; 195:e85-92. [PMID: 17651742 DOI: 10.1016/j.atherosclerosis.2007.06.012] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2007] [Revised: 06/15/2007] [Accepted: 06/19/2007] [Indexed: 11/15/2022]
Abstract
The determinant factors for the development of atherosclerosis in response to dietary cholesterol were examined in two animal models to assess the comparability of results. We studied 128 male Apo E(-/-) and 128LDLr(-/-) mice randomly assigned to baseline (n=8) and 5 groups (n=24 each) that differed only in their dietary fat and cholesterol supplements. At 10, 16, 24 and 32 weeks of age, 8 animals from each group were sequentially sacrificed and the variables analyzed. The lesion sizes changed at different rates but they were predictable and did not differ in complexity. We observed, however, significant differences between strains, particularly in the constitutive expression of liver genes, their metabolic response to dietary cholesterol, their feeding behaviour, their glucose tolerance and the gain in body weight. Both strains presented characteristics that resemble steatohepatitis but manifestations were more severe in LDLr(-/-) mice. The divergent responses indicate that the choice of the diet and the model should be carefully considered in atherosclerosis studies and extrapolations interpreted with caution.
Collapse
Affiliation(s)
- Jorge Joven
- Centre de Recerca Biomèdica, Hospital Universitari de Sant Joan, Carrer Sant Joan s/n, 43201-Reus, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
The opposite-direction modulation of CD4+CD25+ Tregs and T helper 1 cells in acute coronary syndromes. Clin Immunol 2007; 124:90-7. [PMID: 17512253 DOI: 10.1016/j.clim.2007.03.546] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2006] [Revised: 03/19/2007] [Accepted: 03/28/2007] [Indexed: 10/23/2022]
Abstract
Different subsets of T lymphocytes have different functions in atherosclerosis advancement. T helper 1 cells and T regulatory 1 cells have been demonstrated to play opposite roles in rupture of atherosclerotic lesion. However, the role of novel subset of T regulatory cells, known as CD4+CD25+Foxp3+ T cells, remains largely unknown in coronary artery disease (CAD). In this study, we investigated the peripheral CD4+CD25+Foxp3+ T cells of patients with CAD and controls. The patients submitted were divided into three groups: stable angina pectoris (SA) group, unstable angina pectoris (UA) group and acute myocardial infarction (AMI) group. We analyzed the frequencies of peripheral CD4+CD25+Foxp3+ T cells and T helper 1/T helper 2 cells, expression of Foxp3 in CD4+CD25+ T subsets and cytokines pattern in patients and controls. We found that the reduction of CD4+CD25+Foxp3+ T lymphocytes was consistent with the expansion of Th1 cells in patients with unstable CAD. The reversed development between CD4+CD25+ Tregs and Th1 cells might contribute to plaque destabilization.
Collapse
|
48
|
Braunersreuther V, Zernecke A, Arnaud C, Liehn EA, Steffens S, Shagdarsuren E, Bidzhekov K, Burger F, Pelli G, Luckow B, Mach F, Weber C. Ccr5 But Not Ccr1 Deficiency Reduces Development of Diet-Induced Atherosclerosis in Mice. Arterioscler Thromb Vasc Biol 2007; 27:373-9. [PMID: 17138939 DOI: 10.1161/01.atv.0000253886.44609.ae] [Citation(s) in RCA: 223] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Chemokines and their receptors are crucially involved in the development of atherosclerotic lesions by directing monocyte and T cell recruitment. The CC-chemokine receptors 1 (CCR1) and 5 (CCR5) expressed on these cells bind chemokines implicated in atherosclerosis, namely CCL5/RANTES. Although general blockade of CCL5 receptors reduces atherosclerosis, specific roles of CCR1 and CCR5 have not been unequivocally determined. METHODS AND RESULTS We provide two independent lines of investigation to dissect the effects of Ccr1 and Ccr5 deletion in apolipoprotein E-deficient (ApoE-/-) mice in a collaboration between Aachen/Germany and Geneva/Switzerland. Different strains of ApoE-/- Ccr5-/- mice, ApoE-/- Ccr1-/- mice or respective littermates, were fed a high-fat diet for 10 to 12 weeks. Plaque areas were quantified in the aortic roots and thoracoabdominal aortas. Concordantly, both laboratories found that lesion formation was reduced in ApoE-/- Ccr5-/- mice. Plaque quality and immune cells were assessed by immunohistochemistry or mRNA analysis. Whereas lesional macrophage content, aortic CD4, and Th1-related Tim3 expression were reduced, smooth muscle cell (SMC) content and expression of interleukin-10 in plaques, lesional SMCs, and splenocytes were elevated. Protection against lesion formation by Ccr5 deficiency was sustained over 22 weeks of high-fat diet or over 26 weeks of chow diet. Conversely, plaque area, T cell, and interferon-gamma content were increased in ApoE-/- Ccr1-/- mice. CONCLUSIONS Genetic deletion of Ccr5 but not Ccr1 in ApoE-/- mice protects from diet-induced atherosclerosis, associated with a more stable plaque phenotype, reduced mononuclear cell infiltration, Th1-type immune responses, and increased interleukin-10 expression. This corroborates CCR5 as a promising therapeutic target.
Collapse
MESH Headings
- Animals
- Apolipoproteins E/genetics
- Apolipoproteins E/metabolism
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/prevention & control
- Carotid Stenosis/metabolism
- Carotid Stenosis/physiopathology
- Carotid Stenosis/prevention & control
- Cell Proliferation
- Cholesterol, Dietary/adverse effects
- Cytokines/metabolism
- Dietary Fats/adverse effects
- Female
- GATA3 Transcription Factor/genetics
- GATA3 Transcription Factor/metabolism
- Gene Expression Regulation
- Hepatitis A Virus Cellular Receptor 2
- Interleukin-10/genetics
- Interleukin-10/metabolism
- Lymph Nodes/cytology
- Lymph Nodes/metabolism
- Membrane Proteins
- Mice
- Mice, Transgenic
- Phenotype
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, CCR1
- Receptors, CCR5/genetics
- Receptors, CCR5/metabolism
- Receptors, Chemokine/genetics
- Receptors, Chemokine/metabolism
- Receptors, Virus/genetics
- Receptors, Virus/metabolism
- Spleen/cytology
- Spleen/metabolism
- Th1 Cells/cytology
- Th1 Cells/metabolism
Collapse
Affiliation(s)
- Vincent Braunersreuther
- Division of Cardiology, Foundation for Medical Research, Faculty of Medicine, Geneva University Hospital, Geneva, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Diamond GA, Kaul S. From here to eternity: a unified kinetic model for the pathophysiology of atherosclerotic events. Am J Med 2007; 120:5-11. [PMID: 17208070 DOI: 10.1016/j.amjmed.2006.04.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2005] [Accepted: 04/14/2006] [Indexed: 10/23/2022]
Abstract
Two operative pathophysiological models underlie the clinical management of ischemic heart disease: a physical model founded on the magnitude of vascular stenosis and a biochemical model founded on the inflammatory processes within the atherosclerotic plaque. Despite their complementary natures, these 2 models are implicitly competitive--the stenotic model supporting the primacy of aggressive interventional procedures and the inflammatory model supporting the primacy of conservative medical management. We unified these alternative perspectives through a kinetic model that characterizes the pathophysiology of cardiovascular events as a network of exponential transitions between the inflammatory and stenotic states. According to this model, the prevalence of the normative (nonstenotic and noninflammatory) state falls exponentially, while the prevalences of the inflammatory and stenotic states rise to a peak and then fall off exponentially. According to this model, event rate increases as a complex function of both myocardial ischemia and vascular inflammation. Although the model has yet to be prospectively validated, it provides a theoretical foundation for predicting the degree to which atherosclerotic events are due to inflammation versus stenosis and the degree to which they can thereby be prevented by treatment strategies directed at plaque stabilization or relief of ischemia.
Collapse
Affiliation(s)
- George A Diamond
- Division of Cardiology, Cedars-Sinai Medical Center, Los Angeles, Calif, USA.
| | | |
Collapse
|
50
|
Keul P, Tölle M, Lucke S, von Wnuck Lipinski K, Heusch G, Schuchardt M, van der Giet M, Levkau B. The sphingosine-1-phosphate analogue FTY720 reduces atherosclerosis in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol 2006; 27:607-13. [PMID: 17158351 DOI: 10.1161/01.atv.0000254679.42583.88] [Citation(s) in RCA: 130] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The sphingosine-1-phosphate (S1P) analogue FTY720 is a potent immunosuppressive agent currently in Phase III clinical trials for kidney transplantation. FTY720 traps lymphocytes in secondary lymphoid organs thereby preventing their migration to inflammatory sites. Previously, we have identified FTY720 as a potent activator of eNOS. As both inhibition of immune responses and stimulation of eNOS may attenuate atherosclerosis, we administered FTY720 to apolipoprotein E-/- mice fed a high-cholesterol diet. METHODS AND RESULTS FTY720 dramatically reduced atherosclerotic lesion volume (62.5%), macrophage (41.8%), and collagen content (63.5%) after 20 weeks of high-cholesterol diet. In isolated aortic segments and cultured vascular smooth muscle cell, FTY720 potently inhibited thrombin-induced release of monocyte chemoattractant protein-1. This effect was mediated by the S1P3 sphingolipid receptor as FTY720 had no effect on thrombin-induced monocyte chemoattractant protein-1 release in S1P3-/- mice. In contrast to S1P receptors on lymphocytes, FTY720 did not desensitize vascular S1P receptors as arteries from FTY720-treated mice retained their vasodilator response to FTY720-phosphate. CONCLUSIONS We suggest that FTY720 inhibits atherosclerosis by suppressing the machinery involved in monocyte/macrophage emigration to atherosclerotic lesions. As vascular S1P receptors remained functional under FTY720 treatment, S1P agonists that selectively target the vasculature and not the immune system may be promising new drugs against atherosclerosis.
Collapse
MESH Headings
- Animals
- Apolipoproteins E/deficiency
- Atherosclerosis/physiopathology
- Atherosclerosis/prevention & control
- Cell Movement/drug effects
- Cells, Cultured
- Cholesterol, Dietary/administration & dosage
- Cytokines/genetics
- Cytokines/metabolism
- Disease Models, Animal
- Fingolimod Hydrochloride
- Gene Expression Regulation
- Immunohistochemistry
- Lysophospholipids
- Mice
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Probability
- Propylene Glycols/pharmacology
- RNA, Messenger/analysis
- Reverse Transcriptase Polymerase Chain Reaction
- Sphingosine/analogs & derivatives
- Sphingosine/pharmacology
- Statistics, Nonparametric
Collapse
Affiliation(s)
- Petra Keul
- Institute of Pathophysiology, University Hospital Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | | | | | | | | | | | | | | |
Collapse
|