1
|
Azul L, Leandro A, Seiça R, Sena CM. Propagermanium as a Novel Therapeutic Approach for the Treatment of Endothelial Dysfunction in Type 2 Diabetes. Int J Mol Sci 2024; 25:8328. [PMID: 39125901 PMCID: PMC11312737 DOI: 10.3390/ijms25158328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024] Open
Abstract
Propagermanium (PG) has immune modulating activity and anti-inflammatory properties. This work aimed to study the therapeutic efficacy of PG on endothelial and perivascular dysfunction associated with type 2 diabetes. Non-obese type 2 diabetic Goto-Kakizaki (GK) rats were divided into four groups: (1) the control group; (2) the group treated with 50 mg/kg PG; (3) the group fed a high-fat diet (GKHFD); and (4) the group of GKHFD treated with 50 mg/kg PG. PG was given orally for 3 months. Several in vivo parameters and endothelial function were studied in aortas with perivascular adipose tissue PVAT (+) or without PVAT (-). We also determined the vascular inflammation and levels of CD36 in PVAT. In diabetic GK rats, PG did not affect the lipid profile or the results of the intraperitoneal glucose tolerance test. Instead, it improved the fasting glucose levels (18%, p < 0.01), insulin resistance (32%, p < 0.05), endothelial function (33 and 25% in aortas mounted with (+) or without PVAT (-), p < 0.05), and restored the anticontractile effect of the perivascular adipose tissue by reducing its inflammation (56%, p < 0.05) and oxidative stress profile (55%, p < 0.05). Due to its anti-inflammatory characteristics, PG likely improved endothelial dysfunction and restored the perivascular adipose tissue's anticontractile properties.
Collapse
Affiliation(s)
| | | | | | - Cristina M. Sena
- Institute of Physiology, iCBR, Faculty of Medicine, University of Coimbra, Subunit 1, Polo 3, Azinhaga de Santa Comba, Celas, 3000-548 Coimbra, Portugal
| |
Collapse
|
2
|
Wu Y, Ma Y. CCL2-CCR2 signaling axis in obesity and metabolic diseases. J Cell Physiol 2024; 239:e31192. [PMID: 38284280 DOI: 10.1002/jcp.31192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 12/10/2023] [Accepted: 12/29/2023] [Indexed: 01/30/2024]
Abstract
Obesity and metabolic diseases, such as insulin resistance, type 2 diabetes, nonalcoholic fatty liver disease, and cardiovascular ailments, represent formidable global health challenges, bearing considerable implications for both morbidity and mortality rates. It has become increasingly evident that chronic, low-grade inflammation plays a pivotal role in the genesis and advancement of these conditions. The involvement of C-C chemokine ligand 2 (CCL2) and its corresponding receptor, C-C chemokine receptor 2 (CCR2), has been extensively documented in numerous inflammatory maladies. Recent evidence indicates that the CCL2/CCR2 pathway extends beyond immune cell recruitment and inflammation, exerting a notable influence on the genesis and progression of metabolic syndrome. The present review seeks to furnish a comprehensive exposition of the CCL2-CCR2 signaling axis within the context of obesity and metabolic disorders, elucidating its molecular mechanisms, functional roles, and therapeutic implications.
Collapse
Affiliation(s)
- Yue Wu
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Yanchun Ma
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Center for Cell Structure and Function, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| |
Collapse
|
3
|
Li W, Huang Z, Luo Y, Cui Y, Xu M, Luo W, Wu G, Liang G. Tetrandrine alleviates atherosclerosis via inhibition of STING-TBK1 pathway and inflammation in macrophages. Int Immunopharmacol 2023; 119:110139. [PMID: 37099944 DOI: 10.1016/j.intimp.2023.110139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/22/2023] [Accepted: 03/31/2023] [Indexed: 04/28/2023]
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease. Recent studies have showed that stimulator of interferon genes (STING), an important protein in innate immunity, mediates pro-inflammatory activation of macrophages in the development of AS. Tetrandrine (TET) is a natural bisbenzylisoquinoline alkaloid isolated from Stepania tetrandra and possesses anti-inflammatory activities, with unknown effects and mechanisms in AS. In this study, we explored the anti-atherosclerotic effects of TET and investigated the underlying mechanisms. Mouse primary peritoneal macrophages (MPMs) are challenged with cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) or oxidized LDL (oxLDL). We found that pretreatment with TET dose-dependently inhibited cGAMP- or oxLDL-induced STING/ TANK-binding kinase 1 (TBK1) signaling, then suppressing nuclear factor kappa-B (NF-κB) activation and pro-inflammatory factor expression in MPMs. ApoE-/- mice were fed a high-fat diet (HFD) to develop an atherosclerotic phenotype. Administration of TET at 20 mg/kg/day significantly reduced HFD-induced atherosclerotic plaques, accompanied with decreased macrophage infiltration, inflammatory cytokine production, fibrosis, and STING/TBK1 activation in aortic plaque lesions. In summary, we demonstrate that TET inhibits STING/TBK1/NF-κB signaling pathway to reduce inflammation in oxLDL-challenged macrophages and alleviate atherosclerosis in HFD-fed ApoE-/- mice. These findings proved that TET could be a potential therapeutic candidate for the treatment of atherosclerosis-related diseases.
Collapse
Affiliation(s)
- Weixin Li
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zhuqi Huang
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yue Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yaqian Cui
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Mingjiang Xu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Wu Luo
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Gaojun Wu
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Guang Liang
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 311399, China.
| |
Collapse
|
4
|
Zhang H, Yang K, Chen F, Liu Q, Ni J, Cao W, Hua Y, He F, Liu Z, Li L, Fan G. Role of the CCL2-CCR2 axis in cardiovascular disease: Pathogenesis and clinical implications. Front Immunol 2022; 13:975367. [PMID: 36110847 PMCID: PMC9470149 DOI: 10.3389/fimmu.2022.975367] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
The CCL2-CCR2 axis is one of the major chemokine signaling pathways that has received special attention because of its function in the development and progression of cardiovascular disease. Numerous investigations have been performed over the past decades to explore the function of the CCL2-CCR2 signaling axis in cardiovascular disease. Laboratory data on the CCL2-CCR2 axis for cardiovascular disease have shown satisfactory outcomes, yet its clinical translation remains challenging. In this article, we describe the mechanisms of action of the CCL2-CCR2 axis in the development and evolution of cardiovascular diseases including heart failure, atherosclerosis and coronary atherosclerotic heart disease, hypertension and myocardial disease. Laboratory and clinical data on the use of the CCL2-CCR2 pathway as a targeted therapy for cardiovascular diseases are summarized. The potential of the CCL2-CCR2 axis in the treatment of cardiovascular diseases is explored.
Collapse
Affiliation(s)
- Haixia Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Hebei Key Laboratory of Integrated Traditional Chinese and Western Medicine for Diabetes and Its Complications, College of Traditional Chinese Medicine, North China University of Science and Technology, Tangshan, China
| | - Ke Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Feng Chen
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Qianqian Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jingyu Ni
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Weilong Cao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yunqing Hua
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Feng He
- Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources Comprehensive Utilization, Huanggang Normal University, Huanggang, China
| | - Zhihao Liu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lan Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Lan Li, ; Guanwei Fan,
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources Comprehensive Utilization, Huanggang Normal University, Huanggang, China
- *Correspondence: Lan Li, ; Guanwei Fan,
| |
Collapse
|
5
|
Georgakis MK, Bernhagen J, Heitman LH, Weber C, Dichgans M. Targeting the CCL2-CCR2 axis for atheroprotection. Eur Heart J 2022; 43:1799-1808. [PMID: 35567558 DOI: 10.1093/eurheartj/ehac094] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 12/22/2021] [Accepted: 02/15/2022] [Indexed: 11/12/2022] Open
Abstract
Decades of research have established atherosclerosis as an inflammatory disease. Only recently though, clinical trials provided proof-of-concept evidence for the efficacy of anti-inflammatory strategies with respect to cardiovascular events, thus offering a new paradigm for lowering residual vascular risk. Efforts to target the inflammasome-interleukin-1β-interleukin-6 pathway have been highly successful, but inter-individual variations in drug response, a lack of reduction in all-cause mortality, and a higher rate of infections also highlight the need for a second generation of anti-inflammatory agents targeting atherosclerosis-specific immune mechanisms while minimizing systemic side effects. CC-motif chemokine ligand 2/monocyte-chemoattractant protein-1 (CCL2/MCP-1) orchestrates inflammatory monocyte trafficking between the bone marrow, circulation, and atherosclerotic plaques by binding to its cognate receptor CCR2. Adding to a strong body of data from experimental atherosclerosis models, a coherent series of recent large-scale genetic and observational epidemiological studies along with data from human atherosclerotic plaques highlight the relevance and therapeutic potential of the CCL2-CCR2 axis in human atherosclerosis. Here, we summarize experimental and human data pinpointing the CCL2-CCR2 pathway as an emerging drug target in cardiovascular disease. Furthermore, we contextualize previous efforts to interfere with this pathway, scrutinize approaches of ligand targeting vs. receptor targeting, and discuss possible pathway-intrinsic opportunities and challenges related to pharmacological targeting of the CCL2-CCR2 axis in human atherosclerotic disease.
Collapse
Affiliation(s)
- Marios K Georgakis
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, D-81377 Munich, Germany
- Center of Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jürgen Bernhagen
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, D-81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Laura H Heitman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Christian Weber
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Germany
- Institute for Genetic and Biomedical Research, UoS of Milan, National Research Council, Milan, Italy
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, D-81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Munich, Germany
| |
Collapse
|
6
|
Živković L, Asare Y, Bernhagen J, Dichgans M, Georgakis MK. Pharmacological Targeting of the CCL2/CCR2 Axis for Atheroprotection: A Meta-Analysis of Preclinical Studies. Arterioscler Thromb Vasc Biol 2022; 42:e131-e144. [PMID: 35387476 DOI: 10.1161/atvbaha.122.317492] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND The CCL2 (CC-chemokine ligand 2)/CCR2 (CC-chemokine receptor 2) axis governs monocyte recruitment to atherosclerotic lesions. Genetic and epidemiological studies show strong associations of CCL2 levels with atherosclerotic disease. Still, experimental studies testing pharmacological inhibition of CCL2 or CCR2 in atheroprone mice apply widely different approaches and report variable results, thus halting clinical translation. METHODS We systematically searched the literature for studies employing pharmacological CCL2/CCR2 blockade in atheroprone mice and meta-analyzed their effects on lesion size and morphology. RESULTS In a meta-analysis of 14 studies testing 11 different agents, CCL2/CCR2 blockade attenuated atherosclerotic lesion size in the aortic root or arch (g=-0.75 [-1.17 to -0.32], P=6×10-4; N=171/171 mice in experimental/control group), the carotid (g=-2.39 [-4.23 to -0.55], P=0.01; N=24/25), and the femoral artery (g=-2.38 [-3.50 to -1.26], P=3×10-5; N=10/10). Furthermore, CCL2/CCR2 inhibition reduced intralesional macrophage accumulation and increased smooth muscle cell content and collagen deposition. The effects of CCL2/CCR2 inhibition on lesion size correlated with reductions in plaque macrophage accumulation, in accord with a prominent role of CCL2/CCR2 signaling in monocyte recruitment. Subgroup analyses showed comparable efficacy of different CCL2- and CCR2-inhibitors in reducing lesion size and intralesional macrophages. The quality assessment revealed high risk of detection bias due to lack of blinding during outcome assessment, as well as evidence of attrition and reporting bias. CONCLUSIONS Preclinical evidence suggests that pharmacological targeting of CCL2 or CCR2 might lower atherosclerotic lesion burden, but the majority of existing studies suffer major quality issues that highlight the need for additional high-quality research.
Collapse
Affiliation(s)
- Luka Živković
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Germany (L.Ž., Y.A., J.B., M.D., M.K.G.)
| | - Yaw Asare
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Germany (L.Ž., Y.A., J.B., M.D., M.K.G.)
| | - Jürgen Bernhagen
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Germany (L.Ž., Y.A., J.B., M.D., M.K.G.).,Munich Cluster for Systems Neurology (SyNergy), Germany (J.B., M.D.).,Munich Heart Alliance, German Center for Cardiovascular Diseases (DZHK), Germany (J.B.)
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Germany (L.Ž., Y.A., J.B., M.D., M.K.G.).,Munich Cluster for Systems Neurology (SyNergy), Germany (J.B., M.D.).,German Centre for Neurodegenerative Diseases (DZNE), Munich, Germany (M.D.)
| | - Marios K Georgakis
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Germany (L.Ž., Y.A., J.B., M.D., M.K.G.).,Center for Genomic Medicine, Massachusetts General Hospital, Boston (M.K.G.).,Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Boston, MA (M.K.G.)
| |
Collapse
|
7
|
The Entry and Egress of Monocytes in Atherosclerosis: A Biochemical and Biomechanical Driven Process. Cardiovasc Ther 2021; 2021:6642927. [PMID: 34345249 PMCID: PMC8282391 DOI: 10.1155/2021/6642927] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 06/28/2021] [Indexed: 12/24/2022] Open
Abstract
In accordance with “the response to injury” theory, the entry of monocytes into the intima guided by inflammation signals, taking up cholesterol and transforming into foam cells, and egress from plaques determines the progression of atherosclerosis. Multiple cytokines and receptors have been reported to be involved in monocyte recruitment such as CCL2/CCR2, CCL5/CCR5, and CX3CL1/CX3CR1, and the egress of macrophages from the plaque like CCR7/CCL19/CCL21. Interestingly, some neural guidance molecules such as Netrin-1 and Semaphorin 3E have been demonstrated to show an inhibitory effect on monocyte migration. During the processes of monocytes recruitment and migration, factors affecting the biomechanical properties (e.g., the membrane fluidity, the deformability, and stiffness) of the monocytes, like cholesterol, amyloid-β peptide (Aβ), and lipopolysaccharides (LPS), as well as the biomechanical environment that the monocytes are exposed, like the extracellular matrix stiffness, mechanical stretch, blood flow, and hypertension, were discussed in the latter section. Till now, several small interfering RNAs (siRNAs), monoclonal antibodies, and antagonists for CCR2 have been designed and shown promising efficiency on atherosclerosis therapy. Seeking more possible biochemical factors that are chemotactic or can affect the biomechanical properties of monocytes, and uncovering the underlying mechanism, will be helpful in future studies.
Collapse
|
8
|
Blanco-Colio LM, Méndez-Barbero N, Pello Lázaro AM, Aceña Á, Tarín N, Cristóbal C, Martínez-Milla J, González-Lorenzo Ó, Martín-Ventura JL, Huelmos A, Gutiérrez-Landaluce C, López-Castillo M, Kallmeyer A, Cánovas E, Alonso J, López Bescós L, Egido J, Lorenzo Ó, Tuñón J. MCP-1 Predicts Recurrent Cardiovascular Events in Patients with Persistent Inflammation. J Clin Med 2021; 10:jcm10051137. [PMID: 33803115 PMCID: PMC7963189 DOI: 10.3390/jcm10051137] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/23/2021] [Accepted: 03/01/2021] [Indexed: 02/06/2023] Open
Abstract
Clinical data indicate that patients with C-reactive protein (CRP) levels higher than 2 mg per liter suffer from persistent inflammation, which is associated with high risk of cardiovascular disease (CVD). We determined whether a panel of biomarkers associated with CVD could predict recurrent events in patients with low or persistent inflammation and coronary artery disease (CAD). We followed 917 patients with CAD (median 4.59 ± 2.39 years), assessing CRP, galectin-3, monocyte chemoattractant protein-1 (MCP-1), N-terminal fragment of brain natriuretic peptide (NT-proBNP) and troponin-I plasma levels. The primary outcome was the combination of cardiovascular events (acute coronary syndrome, stroke or transient ischemic event, heart failure or death). Patients with persistent inflammation (n = 343) showed higher NT-proBNP and MCP-1 plasma levels compared to patients with CRP < 2 mg/L. Neither MCP-1 nor NT-proBNP was associated with primary outcome in patients with CRP < 2 mg/L. However, NT-proBNP and MCP-1 plasma levels were associated with increased risk of the primary outcome in patients with persistent inflammation. When patients were divided by type of event, MCP-1 was associated with an increased risk of acute ischemic events. A significant interaction between MCP-1 and persistent inflammation was found (synergy index: 6.17 (4.39–7.95)). In conclusion, MCP-1 plasma concentration is associated with recurrent cardiovascular events in patients with persistent inflammation.
Collapse
Affiliation(s)
- Luis M. Blanco-Colio
- Vascular Research Lab, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (L.M.B.-C.); (N.M.-B.); (J.L.M.-V.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Nerea Méndez-Barbero
- Vascular Research Lab, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (L.M.B.-C.); (N.M.-B.); (J.L.M.-V.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ana María Pello Lázaro
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.M.P.L.); (Á.A.); (J.M.-M.); (Ó.G.-L.); (M.L.-C.); (A.K.); (E.C.)
- Department of Medicine, Autónoma University, 08193 Madrid, Spain; (J.E.); (Ó.L.)
| | - Álvaro Aceña
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.M.P.L.); (Á.A.); (J.M.-M.); (Ó.G.-L.); (M.L.-C.); (A.K.); (E.C.)
- Department of Medicine, Autónoma University, 08193 Madrid, Spain; (J.E.); (Ó.L.)
| | - Nieves Tarín
- Department of Cardiology, Hospital Universitario de Móstoles, 28935 Madrid, Spain;
| | - Carmen Cristóbal
- Department of Cardiology, Hospital de Fuenlabrada, 28942 Madrid, Spain; (C.C.); (C.G.-L.)
- Department of Cardiology, Rey Juan Carlos University, Alcorcón, 28933 Madrid, Spain;
| | - Juan Martínez-Milla
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.M.P.L.); (Á.A.); (J.M.-M.); (Ó.G.-L.); (M.L.-C.); (A.K.); (E.C.)
| | - Óscar González-Lorenzo
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.M.P.L.); (Á.A.); (J.M.-M.); (Ó.G.-L.); (M.L.-C.); (A.K.); (E.C.)
| | - José Luis Martín-Ventura
- Vascular Research Lab, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (L.M.B.-C.); (N.M.-B.); (J.L.M.-V.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Medicine, Autónoma University, 08193 Madrid, Spain; (J.E.); (Ó.L.)
| | - Ana Huelmos
- Department of Cardiology, Hospital Universitario Fundación Alcorcón, 28922 Madrid, Spain;
| | | | - Marta López-Castillo
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.M.P.L.); (Á.A.); (J.M.-M.); (Ó.G.-L.); (M.L.-C.); (A.K.); (E.C.)
- Department of Medicine, Autónoma University, 08193 Madrid, Spain; (J.E.); (Ó.L.)
| | - Andrea Kallmeyer
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.M.P.L.); (Á.A.); (J.M.-M.); (Ó.G.-L.); (M.L.-C.); (A.K.); (E.C.)
- Department of Medicine, Autónoma University, 08193 Madrid, Spain; (J.E.); (Ó.L.)
| | - Ester Cánovas
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.M.P.L.); (Á.A.); (J.M.-M.); (Ó.G.-L.); (M.L.-C.); (A.K.); (E.C.)
| | - Joaquín Alonso
- Department of Cardiology, Hospital de Getafe, 28905 Madrid, Spain;
| | - Lorenzo López Bescós
- Department of Cardiology, Rey Juan Carlos University, Alcorcón, 28933 Madrid, Spain;
| | - Jesús Egido
- Department of Medicine, Autónoma University, 08193 Madrid, Spain; (J.E.); (Ó.L.)
- Renal, Vascular and Diabetes Research Lab, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Óscar Lorenzo
- Department of Medicine, Autónoma University, 08193 Madrid, Spain; (J.E.); (Ó.L.)
- Renal, Vascular and Diabetes Research Lab, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José Tuñón
- Vascular Research Lab, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (L.M.B.-C.); (N.M.-B.); (J.L.M.-V.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.M.P.L.); (Á.A.); (J.M.-M.); (Ó.G.-L.); (M.L.-C.); (A.K.); (E.C.)
- Department of Medicine, Autónoma University, 08193 Madrid, Spain; (J.E.); (Ó.L.)
- Correspondence:
| |
Collapse
|
9
|
Georgakis MK, Malik R, Björkbacka H, Pana TA, Demissie S, Ayers C, Elhadad MA, Fornage M, Beiser A, Benjamin EJ, Boekholdt MS, Engström G, Herder C, Hoogeveen RC, Koenig W, Melander O, Orho-Melander M, Schiopu A, Söderholm M, Wareham N, Ballantyne CM, Peters A, Seshadri S, Myint PK, Nilsson J, de Lemos JA, Dichgans M. Circulating Monocyte Chemoattractant Protein-1 and Risk of Stroke: Meta-Analysis of Population-Based Studies Involving 17 180 Individuals. Circ Res 2019; 125:773-782. [PMID: 31476962 PMCID: PMC6763364 DOI: 10.1161/circresaha.119.315380] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 09/01/2019] [Indexed: 12/13/2022]
Abstract
Rationale: Proinflammatory cytokines have been identified as potential targets for lowering vascular risk. Experimental evidence and Mendelian randomization suggest a role of MCP-1 (monocyte chemoattractant protein-1) in atherosclerosis and stroke. However, data from large-scale observational studies are lacking. Objective: To determine whether circulating levels of MCP-1 are associated with risk of incident stroke in the general population. Methods and Results: We used previously unpublished data on 17 180 stroke-free individuals (mean age, 56.7±8.1 years; 48.8% men) from 6 population-based prospective cohort studies and explored associations between baseline circulating MCP-1 levels and risk of any stroke, ischemic stroke, and hemorrhagic stroke during a mean follow-up interval of 16.3 years (280 522 person-years at risk; 1435 incident stroke events). We applied Cox proportional-hazards models and pooled hazard ratios (HRs) using random-effects meta-analyses. After adjustments for age, sex, race, and vascular risk factors, higher MCP-1 levels were associated with increased risk of any stroke (HR per 1-SD increment in ln-transformed MCP-1, 1.07; 95% CI, 1.01-1.14). Focusing on stroke subtypes, we found a significant association between baseline MCP-1 levels and higher risk of ischemic stroke (HR, 1.11 [1.02-1.21]) but not hemorrhagic stroke (HR, 1.02 [0.82-1.29]). The results followed a dose-response pattern with a higher risk of ischemic stroke among individuals in the upper quartiles of MCP-1 levels as compared with the first quartile (HRs, second quartile: 1.19 [1.00-1.42]; third quartile: 1.35 [1.14-1.59]; fourth quartile: 1.38 [1.07-1.77]). There was no indication for heterogeneity across studies, and in a subsample of 4 studies (12 516 individuals), the risk estimates were stable after additional adjustments for circulating levels of IL (interleukin)-6 and high-sensitivity CRP (C-reactive protein). Conclusions: Higher circulating levels of MCP-1 are associated with increased long-term risk of stroke. Our findings along with genetic and experimental evidence suggest that MCP-1 signaling might represent a therapeutic target to lower stroke risk.Visual Overview: An online visual overview is available for this article.
Collapse
Affiliation(s)
- Marios K. Georgakis
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University LMU, Munich
- Graduate School for Systemic Neurosciences (GSN), Ludwig-Maximilians-University LMU, Munich
| | - Rainer Malik
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University LMU, Munich
| | | | - Tiberiu Alexandru Pana
- Institute of Applied Health Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen
| | - Serkalem Demissie
- Biostatistics, Boston University School of Public Health, Boston MA
- National Heart, Lung, and Blood Institute’s and Boston University’s Framingham Heart Study, Framingham, MA
| | - Colby Ayers
- Cardiology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Mohamed A. Elhadad
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg
- German Research Center for Cardiovascular Disease (DZHK), Partner site Munich Heart Alliance, Munich, Germany
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School and Human Genetics Center, School of Public Health, University of Texas Health Science Center, Houston, TX
| | - Alexa Beiser
- Biostatistics, Boston University School of Public Health, Boston MA
- National Heart, Lung, and Blood Institute’s and Boston University’s Framingham Heart Study, Framingham, MA
- Neurology, Boston University School of Medicine
| | - Emelia J. Benjamin
- National Heart, Lung, and Blood Institute’s and Boston University’s Framingham Heart Study, Framingham, MA
- Medicine, Boston University School of Medicine
- Epidemiology, Boston University School of Public Health
| | | | | | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf
- German Center for Diabetes Research (DZD), Partner Düsseldorf
- Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Düsseldorf
| | | | - Wolfgang Koenig
- German Research Center for Cardiovascular Disease (DZHK), Partner site Munich Heart Alliance, Munich, Germany
- Deutsches Herzzentrum München, Technische Universität München
- Institute of Epidemiology and Biostatistics, University of Ulm, Ulm, Germany
| | | | | | - Alexandru Schiopu
- Clinical Sciences, Malmö, Lund University, Malmö
- Cardiology, Skåne University Hospital, Malmö, Sweden
| | | | - Nick Wareham
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | | | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg
| | - Sudha Seshadri
- National Heart, Lung, and Blood Institute’s and Boston University’s Framingham Heart Study, Framingham, MA
- Medicine, Boston University School of Medicine
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio
| | - Phyo K. Myint
- Institute of Applied Health Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen
| | - Jan Nilsson
- Clinical Sciences, Malmö, Lund University, Malmö
| | - James A. de Lemos
- Cardiology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University LMU, Munich
- Munich Cluster for Systems Neurology (SyNergy)
- German Centre for Neurodegenerative Diseases (DZNE)
| |
Collapse
|
10
|
Yumimoto K, Sugiyama S, Mimori K, Nakayama KI. Potentials of C-C motif chemokine 2-C-C chemokine receptor type 2 blockers including propagermanium as anticancer agents. Cancer Sci 2019; 110:2090-2099. [PMID: 31111571 PMCID: PMC6609805 DOI: 10.1111/cas.14075] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/09/2019] [Accepted: 05/14/2019] [Indexed: 12/21/2022] Open
Abstract
Inflammation plays an essential role in the development and progression of most cancers. Chemokine C‐C motif chemokine 2 (CCL2) and its receptor C‐C chemokine receptor type 2 (CCR2) constitute a key signaling axis in inflammation that has recently attracted much interest on the basis of evidence showing its association with cancer progression. Propagermanium (3‐oxygermylpropionic acid polymer) is an organogermanium compound that is given for the treatment of hepatitis B in Japan and which inhibits the CCL2‐CCR2 signaling pathway. Herein, we review the importance of the CCL2‐CCR2 axis as a target in cancer treatment as shown by studies in mice and humans with pharmacological agents including propagermanium.
Collapse
Affiliation(s)
- Kanae Yumimoto
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Shigeaki Sugiyama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Koshi Mimori
- Department of Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| |
Collapse
|
11
|
Lawal IO, Ankrah AO, Stoltz AC, Sathekge MM. Radionuclide imaging of inflammation in atherosclerotic vascular disease among people living with HIV infection: current practice and future perspective. Eur J Hybrid Imaging 2019; 3:5. [PMID: 34191183 PMCID: PMC8218042 DOI: 10.1186/s41824-019-0053-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/11/2019] [Indexed: 01/03/2023] Open
Abstract
People living with human immunodeficiency virus (HIV) infection have twice the risk of atherosclerotic vascular disease compared with non-infected individuals. Inflammation plays a critical role in the development and progression of atherosclerotic vascular disease. Therapies targeting inflammation irrespective of serum lipid levels have been shown to be effective in preventing the occurrence of CVD. Radionuclide imaging is a viable method for evaluating arterial inflammation. This evaluation is useful in quantifying CVD risk and for assessing the effectiveness of anti-inflammatory treatment. The most tested radionuclide method for quantifying arterial inflammation among people living with HIV infection has been with F-18 FDG PET/CT. The level of arterial uptake of F-18 FDG correlates with vascular inflammation and with the risk of development and progression of atherosclerotic disease. Several limitations exist to the use of F-18 FDG for PET quantification of arterial inflammation. Many targets expressed on macrophage, a significant player in arterial inflammation, have the potential for use in evaluating arterial inflammation among people living with HIV infection. The review describes the clinical utility of F-18 FDG PET/CT in assessing arterial inflammation as a risk for atherosclerotic disease among people living with HIV infection. It also outlines potential newer probes that may quantify arterial inflammation in the HIV-infected population by targeting different proteins expressed on macrophages.
Collapse
Affiliation(s)
- Ismaheel O. Lawal
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Private Bag X169, Pretoria, 0001 South Africa
| | - Alfred O. Ankrah
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Private Bag X169, Pretoria, 0001 South Africa
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen & University of Groningen, Groningen, The Netherlands
| | - Anton C. Stoltz
- Infectious Disease Unit, Department of Internal Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria, South Africa
| | - Mike M. Sathekge
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Private Bag X169, Pretoria, 0001 South Africa
| |
Collapse
|
12
|
Monocyte subtypes and the CCR2 chemokine receptor in cardiovascular disease. Clin Sci (Lond) 2017; 131:1215-1224. [DOI: 10.1042/cs20170009] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 02/17/2017] [Accepted: 02/24/2017] [Indexed: 12/14/2022]
Abstract
Monocytes circulate in the blood and migrate to inflammatory tissues, but their functions can be either detrimental or beneficial, depending on their phenotypes. In humans, classical monocytes are inflammatory cluster of differentiation (CD)14++CD16−CCR2++ cells originated from the bone marrow or spleen reservoirs and comprise ≥92% of monocytes. Intermediate monocytes (CD14++CD16+CCR2+) are involved in the production of anti-inflammatory cytokines [such as interleukin (IL)-10], reactive oxygen species (ROS), and proinflammatory mediators [such as tumor necrosis factor-α (TNF-α) and IL-1β). Nonclassical monocytes (CD14+CD16++CCR2−) are patrolling cells involved in tissue repair and debris removal from the vasculature. Many studies in both humans and animals have shown the importance of monocyte chemoattractant protein-1 (MCP-1) and its receptor [chemokine receptor of MCP-1 (CCR2)] in pathologies, such as atherosclerosis and myocardial infarction (MI). This review presents the importance of these monocyte subsets in cardiovascular diseases (CVDs), and sheds light on new strategies for the blocking of the MCP-1/CCR2 axis as a therapeutic goal for treating vascular disorders.
Collapse
|
13
|
The CCR2 Inhibitor Propagermanium Attenuates Diet-Induced Insulin Resistance, Adipose Tissue Inflammation and Non-Alcoholic Steatohepatitis. PLoS One 2017; 12:e0169740. [PMID: 28076416 PMCID: PMC5226841 DOI: 10.1371/journal.pone.0169740] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 12/21/2016] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND AND AIM Obese patients with chronic inflammation in white adipose tissue (WAT) have an increased risk of developing non-alcoholic steatohepatitis (NASH). The C-C chemokine receptor-2 (CCR2) has a crucial role in the recruitment of immune cells to WAT and liver, thereby promoting the inflammatory component of the disease. Herein, we examined whether intervention with propagermanium, an inhibitor of CCR2, would attenuate tissue inflammation and NASH development. METHODS Male C57BL/6J mice received a high-fat diet (HFD) for 0, 6, 12 and 24 weeks to characterize the development of early disease symptoms of NASH, i.e. insulin resistance and WAT inflammation (by hyperinsulinemic-euglycemic clamp and histology, respectively) and to define the optimal time point for intervention. In a separate study, mice were pretreated with HFD followed by propagermanium treatment (0.05% w/w) after 6 weeks (early intervention) or 12 weeks (late intervention). NASH was analyzed after 24 weeks of diet feeding. RESULTS Insulin resistance in WAT developed after 6 weeks of HFD, which was paralleled by modest WAT inflammation. Insulin resistance and inflammation in WAT intensified after 12 weeks of HFD, and preceded NASH development. The subsequent CCR2 intervention experiment showed that early, but not late, propagermanium treatment attenuated insulin resistance. Only the early treatment significantly decreased Mcp-1 and CD11c gene expression in WAT, indicating reduced WAT inflammation. Histopathological analysis of liver demonstrated that propagermanium treatment decreased macrovesicular steatosis and tended to reduce lobular inflammation, with more pronounced effects in the early intervention group. Propagermanium improved the ratio between pro-inflammatory (M1) and anti-inflammatory (M2) macrophages, quantified by CD11c and Arginase-1 gene expression in both intervention groups. CONCLUSIONS Overall, early propagermanium administration was more effective to improve insulin resistance, WAT inflammation and NASH compared to late intervention. These data suggest that therapeutic interventions for NASH directed at the MCP-1/CCR2 pathway should be initiated early.
Collapse
|
14
|
Fujimura N, Xu B, Dalman J, Deng H, Aoyama K, Dalman RL. CCR2 inhibition sequesters multiple subsets of leukocytes in the bone marrow. Sci Rep 2015. [PMID: 26206182 PMCID: PMC4513281 DOI: 10.1038/srep11664] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Chemokine receptor CCR2 mediates monocyte mobilization from the bone marrow (BM) and subsequent migration into target tissues. The degree to which CCR2 is differentially expressed in leukocyte subsets, and the contribution of CCR2 to these leukocyte mobilization from the BM are poorly understood. Using red fluorescence protein CCR2 reporter mice, we found heterogeneity in CCR2 expression among leukocyte subsets in varying tissues. CCR2 was highly expressed by inflammatory monocytes, dendritic cells, plasmacytoid dendritic cells and NK cells in all tissues. Unexpectedly, more than 60% of neutrophils expressed CCR2, albeit at low levels. CCR2 expression in T cells, B cells and NK T cells was greatest in the BM compared to other tissues. Genetic CCR2 deficiency markedly sequestered all leukocyte subsets in the BM, with reciprocal reduction noted in the peripheral blood and spleen. CCR2 inhibition via treatment with CCR2 signaling inhibitor propagermanium produced similar effects. Propagermanium also mitigated lipopolysaccharide-induced BM leukocyte egress. Consistent with its functional significance, CCR2 antibody staining revealed surface CCR2 expression within a subset of BM neutrophils. These results demonstrate the central role CCR2 plays in mediating leukocyte mobilization from the BM, and suggest a role for CCR2 inhibition in managing monocytes/macrophages-mediated chronic inflammatory conditions.
Collapse
Affiliation(s)
- Naoki Fujimura
- 1] Departments of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA [2] Department of Vascular Surgery, Saiseikai Central Hospital, Minato-Ku Mita 1-4-17, Tokyo 108-0073, Japan
| | - Baohui Xu
- Departments of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jackson Dalman
- Departments of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hongping Deng
- Departments of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kohji Aoyama
- Department of Hygiene and Health Promotion Medicine, Kagoshima University School of Medicine, Sakuragaoka 8-35-1, Kagoshima 890-0075, Japan
| | - Ronald L Dalman
- Departments of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
15
|
Gomes Quinderé AL, Benevides NMB, Carbone F, Mach F, Vuilleumier N, Montecucco F. Update on selective treatments targeting neutrophilic inflammation in atherogenesis and atherothrombosis. Thromb Haemost 2013; 111:634-46. [PMID: 24285257 DOI: 10.1160/th13-08-0712] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 10/28/2013] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is the most common pathological process underlying cardiovascular diseases. Current therapies are largely focused on alleviating hyperlipidaemia and preventing thrombotic complications, but do not completely eliminate risk of suffering recurrent acute ischaemic events. Specifically targeting the inflammatory processes may help to reduce this residual risk of major adverse cardiovascular events in atherosclerotic patients. The involvement of neutrophils in the pathophysiology of atherosclerosis is an emerging field, where evidence for their causal contribution during various stages of atherosclerosis is accumulating. Therefore, the identification of neutrophils as a potential therapeutic target may offer new therapeutic perspective to reduce the current atherosclerotic burden. This narrative review highlights the expanding role of neutrophils in atherogenesis and discusses on the potential treatment targeting neutrophil-related inflammation and associated atherosclerotic plaque vulnerability.
Collapse
Affiliation(s)
| | | | | | | | | | - Fabrizio Montecucco
- Fabrizio Montecucco, MD, PhD, Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, 4 rue Gabrielle-Perret-Gentil, 1205 Geneva, Switzerland, Tel: +41 22 38 27 238, Fax: +41 22 38 27 245, E mail:
| |
Collapse
|
16
|
Martinez HG, Quinones MP, Jimenez F, Estrada C, Clark KM, Suzuki K, Miura N, Ohno N, Ahuja SK, Ahuja SS. Important role of CCR2 in a murine model of coronary vasculitis. BMC Immunol 2012; 13:56. [PMID: 23074996 PMCID: PMC3519555 DOI: 10.1186/1471-2172-13-56] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 10/09/2012] [Indexed: 12/24/2022] Open
Abstract
Background Chemokines and their receptors play a role in the innate immune response as well as in the disruption of the balance between pro-inflammatory Th17 cells and regulatory T cells (Treg), underlying the pathogenesis of coronary vasculitis in Kawasaki disease (KD). Results Here we show that genetic inactivation of chemokine receptor (CCR)-2 is protective against the induction of aortic and coronary vasculitis following injection of Candida albicans water-soluble cell wall extracts (CAWS). Mechanistically, both T and B cells were required for the induction of vasculitis, a role that was directly modulated by CCR2. CAWS administration promoted mobilization of CCR2-dependent inflammatory monocytes (iMo) from the bone marrow (BM) to the periphery as well as production of IL-6. IL-6 was likely to contribute to the depletion of Treg and expansion of Th17 cells in CAWS-injected Ccr2+/+ mice, processes that were ameliorated following the genetic inactivation of CCR2. Conclusion Collectively, our findings provide novel insights into the role of CCR2 in the pathogenesis of vasculitis as seen in KD and highlight novel therapeutic targets, specifically for individuals resistant to first-line treatments.
Collapse
Affiliation(s)
- Hernan G Martinez
- Department of Medicine (MC 7870), University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Vaitilingam B, Chelvam V, Kularatne SA, Poh S, Ayala-Lopez W, Low PS. A Folate Receptor-α–Specific Ligand That Targets Cancer Tissue and Not Sites of Inflammation. J Nucl Med 2012; 53:1127-34. [DOI: 10.2967/jnumed.111.099390] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
18
|
Gratchev A, Sobenin I, Orekhov A, Kzhyshkowska J. Monocytes as a diagnostic marker of cardiovascular diseases. Immunobiology 2012; 217:476-82. [PMID: 22325375 DOI: 10.1016/j.imbio.2012.01.008] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2012] [Accepted: 01/07/2012] [Indexed: 02/07/2023]
Abstract
Inflammation is an important pathogenic factor of cardiovascular diseases. Inflammatory processes induce the organism systemic changes that are sensed by the cells of innate immune system. These systemic changes include increased concentrations of soluble factors capable of activating monocytes in the blood circulation therefore monocytes represent highly attractive cell population for diagnostic use. To date various parameters of circulating monocytes were associated with cardiovascular diseases. These comprise monocyte count, increased adhesive properties, alteration of lipid metabolism, phagocytosis and endocytosis of LDL. Search for markers, better suitable for clinical use led to identification of monocyte population heterogeneity. One of the best studied markers for identification of monocyte subpopulation is CD16. Although there is no consensus regarding the origin and composition of various monocyte subpopulations, association of increased size of CD16+ monocyte population with atherosclerosis is well established. Further surface markers of monocytes found to be associated with cardiovascular diseases are CD18, CD11b, CXCR1, CD36 and STAB1. Functional studies performed on primary human monocytes support the importance of these molecules for the pathogenesis of cardiovascular diseases. Continuous research on monocyte biology leads to identification of perspective markers that show significant potential of clinical use. These include analysis of monocyte response to a challenge and level of mitochondrial DNA heteroplasmy. Further research involving genomic, proteomic and cell biology techniques supplemented with systems biology approaches for data analysis and computer simulations are required for defining molecular and functional parameters of monocytes to be used as a diagnostic tool or therapeutic target.
Collapse
Affiliation(s)
- Alexei Gratchev
- Department of Dermatology, University Medical Center and Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg, Mannheim, Germany.
| | | | | | | |
Collapse
|
19
|
Okamoto M, Fuchigami M, Suzuki T, Watanabe N. A Novel C–C Chemokine Receptor 2 Antagonist Prevents Progression of Albuminuria and Atherosclerosis in Mouse Models. Biol Pharm Bull 2012; 35:2069-74. [DOI: 10.1248/bpb.b12-00528] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
20
|
Kang YS, Cha JJ, Hyun YY, Cha DR. Novel C-C chemokine receptor 2 antagonists in metabolic disease: a review of recent developments. Expert Opin Investig Drugs 2011; 20:745-56. [PMID: 21466412 DOI: 10.1517/13543784.2011.575359] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION C-C chemokine ligand 2 (CCL2), also known as monocyte chemoattractant protein-1, and its receptor, C-C chemokine receptor 2 (CCR2), play important roles in various inflammatory diseases. Recently, it has been reported that the CCL2/CCR2 pathway also has an important role in the pathogenesis of metabolic syndrome through its association with obesity and related systemic complications. AREAS COVERED This review focuses on the roles of CCR2 in the pathogenesis of adipose tissue inflammation and other organ damage associated with metabolic syndrome, which is still a matter of debate in many studies. It also covers the use of novel CCR2 antagonists as therapies in such conditions. EXPERT OPINION There is abundant experimental evidence that the CCL2/CCR2 pathway may be involved in chronic low-grade inflammation of adipose tissue in obesity and related metabolic diseases. Although animal models of diabetes and obesity, as well as human trials, have produced controversial results, there is continued interest in the roles of CCR2 inhibition in metabolic disease. Further identification of the mechanisms for recruitment and activation of phagocytes and determination of the roles of other chemokines are needed. Future study of these fundamental questions will provide a clearer understanding of adipose tissue biology and potential therapeutic targets for treatment of obesity-related metabolic disease, including diabetic nephropathy.
Collapse
Affiliation(s)
- Young Sun Kang
- Medical College of Korea University, Ansan Hospital, Division of Nephrology, Department of Internal Medicine, Ansan City, Korea
| | | | | | | |
Collapse
|
21
|
Choi SE, Jang HJ, Kang Y, Jung JG, Han SJ, Kim HJ, Kim DJ, Lee KW. Atherosclerosis induced by a high-fat diet is alleviated by lithium chloride via reduction of VCAM expression in ApoE-deficient mice. Vascul Pharmacol 2010; 53:264-72. [PMID: 20888430 DOI: 10.1016/j.vph.2010.09.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2010] [Revised: 08/20/2010] [Accepted: 09/24/2010] [Indexed: 10/19/2022]
Abstract
Endothelial cell dysfunction may play an important role in the development of various vascular diseases, including atherosclerosis. Here we investigated whether lithium chloride (LiCl), an inhibitor of glycogen synthase kinase-3β (GSK-3β), could counteract atherosclerosis induced by a high-fat diet in ApoE⁻/⁻ mice. Ten-week-old male mice were randomly divided into four groups: normal chow diet, high-fat diet (i.e., 20% fat and 0.5% cholesterol), high-fat diet with LiCl treatment for 6 weeks and high-fat diet with LiCl treatment for 14 weeks. Examination of plasma profiles indicated that blood glucose levels were significantly decreased by LiCl treatment. Supplementation with LiCl dramatically reduced atherosclerotic lesion formation in the aorta and aortic root. LiCl treatment also decreased vascular cell adhesion molecule (VCAM)-1 expression and macrophage infiltration into atherosclerotic lesion areas within the aortic valve. In addition, inhibition of GSK-3β by TDZD-8, SB216763, and LiCl, as well as adenoviral transduction with a catalytically inactive GSK-3β, reduced palmitate-induced VCAM-1 expression through inhibition of JNK activity and degradation of Iκ-Bα in human umbilical vein endothelial cells (HUVECs). The results of the present study suggest that LiCl alleviates palmitate-induced cell adhesion molecule expression in HUVECs and decreases atherosclerosis induced by a high-fat diet in ApoE⁻/⁻ mice. Thus, GSK-3β may be involved in the development of atherosclerosis induced by a high-fat diet in ApoE⁻/⁻ mice.
Collapse
Affiliation(s)
- Sung-E Choi
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Kyunggi-do 442-749, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Zhao Q. Dual targeting of CCR2 and CCR5: therapeutic potential for immunologic and cardiovascular diseases. J Leukoc Biol 2010; 88:41-55. [PMID: 20360402 DOI: 10.1189/jlb.1009671] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
A cardinal feature of inflammation is the tissue recruitment of leukocytes, a process that is mediated predominantly by chemokines via their receptors on migrating cells. CCR2 and CCR5, two CC chemokine receptors, are important players in the trafficking of monocytes/macrophages and in the functions of other cell types relevant to disease pathogenesis. This review provides a brief overview of the biological actions of CCR2 and CCR5 and a comprehensive summary of published data that demonstrate the involvement of both receptors in the pathogenesis of immunologic diseases (RA, CD, and transplant rejection) and cardiovascular diseases (atherosclerosis and AIH). In light of the potential for functional redundancy of chemokine receptors in mediating leukocyte trafficking and the consequent concern over insufficient efficacy offered by pharmacologically inhibiting one receptor, this review presents evidence supporting dual targeting of CCR2 and CCR5 as a more efficacious strategy than targeting either receptor alone. It also examines potential safety issues associated with such dual targeting.
Collapse
Affiliation(s)
- Qihong Zhao
- Research and Development, Bristol-Myers Squibb, Princeton, New Jersey 08543, USA.
| |
Collapse
|
23
|
Tamura Y, Sugimoto M, Murayama T, Minami M, Nishikaze Y, Ariyasu H, Akamizu T, Kita T, Yokode M, Arai H. C-C chemokine receptor 2 inhibitor improves diet-induced development of insulin resistance and hepatic steatosis in mice. J Atheroscler Thromb 2010; 17:219-28. [PMID: 20179360 DOI: 10.5551/jat.3368] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
AIM Adipose tissue inflammation induced by macrophage infiltration through the MCP-1/CCR2 pathway is considered to play a pivotal role in the development of visceral obesity and insulin resistance. In the present study, therefore, we examined whether pharmacological inhibition of CCR2 is effective against the development of diet-induced metabolic disorders. METHODS C57BL/6 mice were fed a high fat and sucrose diet with or without propagermanium (CCR2 inhibitor, 5 or 50 mg/kg BW/day) for 12 weeks from 6 weeks of age. Then we analyzed lipid and glucose metabolism and tissue inflammation in the liver and adipose tissues along with serum markers in those mice. RESULTS AND CONCLUSION Propagermanium treatment slightly decreased body weight gain and visceral fat accumulation in diet-induced obese (DIO) mice. Further, propagermanium suppressed macrophage accumulation and shifted adipose tissue macrophage polarization from the pro-inflammatory (M1) state to anti-inflammatory (M2) state in DIO mice. Expressions of TNF-alpha and MCP-1 mRNA in adipose tissue were reduced by propagermanium treatment, indicating that propagermanim suppressed inflammation in adipose tissue. Propagermanium treatment also ameliorated glucose tolerance, insulin sensitivity, and decreased hepatic triglyceride in DIO mice. Thus, propagermanium improved diet-induced obesity and related metabolic disorders, such as insulin resistance and hepatic steatosis by suppressing inflammation in adipose tissue. Our data indicate that inhibition of CCR2 could improve diet-induced metabolic disorders, and that propagermanium may be a beneficial drug for the treatment of metabolic syndrome.
Collapse
Affiliation(s)
- Yukinori Tamura
- Department of Clinical Innovative Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Olzinski AR, Turner GH, Bernard RE, Karr H, Cornejo CA, Aravindhan K, Hoang B, Ringenberg MA, Qin P, Goodman KB, Willette RN, Macphee CH, Jucker BM, Sehon CA, Gough PJ. Pharmacological inhibition of C-C chemokine receptor 2 decreases macrophage infiltration in the aortic root of the human C-C chemokine receptor 2/apolipoprotein E-/- mouse: magnetic resonance imaging assessment. Arterioscler Thromb Vasc Biol 2009; 30:253-9. [PMID: 19965779 DOI: 10.1161/atvbaha.109.198812] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
UNLABELLED Purpose- This study assessed the pharmacological effect of a novel selective C-C chemokine receptor (CCR) 2 antagonist (GSK1344386B) on monocyte/macrophage infiltration into atherosclerotic plaque using magnetic resonance imaging (MRI) in an atherosclerotic mouse model. METHODS AND RESULTS Apolipoprotein E(-/-) mice expressing human CCR2 were fed a Western diet (vehicle group) or a Western diet plus10 mg/kg per day of GSK1344386B (GSK1344386B group). After the baseline MRI, mice were implanted with osmotic pumps containing angiotensin II, 1000 ng/kg per minute, to accelerate lesion formation. After five weeks of angiotensin II administration, mice received ultrasmall superparamagnetic iron oxide, an MRI contrast agent for the assessment of monocyte/macrophage infiltration to the plaque, and underwent imaging. After imaging, mice were euthanized, and the heart and aorta were harvested for ex vivo MRI and histopathological examination. After 5 weeks of dietary dosing, there were no significant differences between groups in body or liver weight or plasma cholesterol concentrations. An in vivo MRI reflected a decrease in ultrasmall superparamagnetic iron oxide contrast agent uptake in the aortic arch of the GSK1344386B group (P<0.05). An ex vivo MRI of the aortic root also reflected decreased ultrasmall superparamagnetic iron oxide uptake in the GSK1344386B group and was verified by absolute iron analysis (P<0.05). Although there was no difference in aortic root lesion area between groups, there was a 30% reduction in macrophage area observed in the GSK1344386B group (P<0.05). CONCLUSIONS An MRI was used to noninvasively assess the decreased macrophage content in the atherosclerotic plaque after selective CCR2 inhibition.
Collapse
Affiliation(s)
- Alan R Olzinski
- Cardiovascular and Urogenital Center of Excellence in Drug Discovery, King of Prussia, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Rotzius P, Soehnlein O, Kenne E, Lindbom L, Nystrom K, Thams S, Eriksson EE. ApoE−/−/Lysozyme MEGFP/EGFP mice as a versatile model to study monocyte and neutrophil trafficking in atherosclerosis. Atherosclerosis 2009; 202:111-8. [DOI: 10.1016/j.atherosclerosis.2008.04.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2007] [Revised: 03/26/2008] [Accepted: 04/08/2008] [Indexed: 10/22/2022]
|
26
|
Tamura Y, Sugimoto M, Murayama T, Ueda Y, Kanamori H, Ono K, Ariyasu H, Akamizu T, Kita T, Yokode M, Arai H. Inhibition of CCR2 Ameliorates Insulin Resistance and Hepatic Steatosis in db/db Mice. Arterioscler Thromb Vasc Biol 2008; 28:2195-201. [DOI: 10.1161/atvbaha.108.168633] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Objective—
Recently, adipose tissue inflammation induced by macrophage infiltration through MCP-1/C-C chemokine receptor-2 (CCR2) pathway is considered to play a role in the development of visceral obesity and insulin resistance. In the present study, to further examine the role of CCR2 in the development of obesity and type 2 diabetes, we studied the effect of pharmacological inhibition of CCR2 from the early stage of obesity in db/db mice.
Methods and Results—
Db/+m (lean control) and db/db mice were fed with a standard diet with or without 0.005% propagermanium, as a CCR2 inhibitor for 12 weeks from 6 weeks of age. Propagermanium treatment decreased body weight gain, visceral fat accumulation, and the size of adipocytes only in db/db mice. Further, propagermanium suppressed macrophage accumulation and inflammation in adipose tissue. Propagermanium treatment also ameliorated glucose tolerance and insulin sensitivity, and decreased hepatic triglyceride contents in db/db mice.
Conclusions—
Propagermanium improved obesity and related metabolic disorders, such as insulin resistance and hepatic steatosis by suppressing inflammation in adipose tissue. Our data indicate that inhibition of CCR2 could improve obesity and type 2 diabetes by interfering adipose tissue inflammation, and that propagermanium may be a beneficial drug for the treatment of the metabolic syndrome.
Collapse
Affiliation(s)
- Yukinori Tamura
- From the Departments of Clinical Innovative Medicine (Y.T., M.S., T.M., M.Y.), Cardiovascular Medicine (Y.U., K.O., T.K.), Nephrology (H.K.), and Geriatric Medicine (H.A.), Kyoto University Graduate School of Medicine (H.A., T.A.), Translational Research Center, Kyoto University Hospital (H.A., T.A.), Japan
| | - Masayuki Sugimoto
- From the Departments of Clinical Innovative Medicine (Y.T., M.S., T.M., M.Y.), Cardiovascular Medicine (Y.U., K.O., T.K.), Nephrology (H.K.), and Geriatric Medicine (H.A.), Kyoto University Graduate School of Medicine (H.A., T.A.), Translational Research Center, Kyoto University Hospital (H.A., T.A.), Japan
| | - Toshinori Murayama
- From the Departments of Clinical Innovative Medicine (Y.T., M.S., T.M., M.Y.), Cardiovascular Medicine (Y.U., K.O., T.K.), Nephrology (H.K.), and Geriatric Medicine (H.A.), Kyoto University Graduate School of Medicine (H.A., T.A.), Translational Research Center, Kyoto University Hospital (H.A., T.A.), Japan
| | - Yukihiko Ueda
- From the Departments of Clinical Innovative Medicine (Y.T., M.S., T.M., M.Y.), Cardiovascular Medicine (Y.U., K.O., T.K.), Nephrology (H.K.), and Geriatric Medicine (H.A.), Kyoto University Graduate School of Medicine (H.A., T.A.), Translational Research Center, Kyoto University Hospital (H.A., T.A.), Japan
| | - Hiroshi Kanamori
- From the Departments of Clinical Innovative Medicine (Y.T., M.S., T.M., M.Y.), Cardiovascular Medicine (Y.U., K.O., T.K.), Nephrology (H.K.), and Geriatric Medicine (H.A.), Kyoto University Graduate School of Medicine (H.A., T.A.), Translational Research Center, Kyoto University Hospital (H.A., T.A.), Japan
| | - Koh Ono
- From the Departments of Clinical Innovative Medicine (Y.T., M.S., T.M., M.Y.), Cardiovascular Medicine (Y.U., K.O., T.K.), Nephrology (H.K.), and Geriatric Medicine (H.A.), Kyoto University Graduate School of Medicine (H.A., T.A.), Translational Research Center, Kyoto University Hospital (H.A., T.A.), Japan
| | - Hiroyuki Ariyasu
- From the Departments of Clinical Innovative Medicine (Y.T., M.S., T.M., M.Y.), Cardiovascular Medicine (Y.U., K.O., T.K.), Nephrology (H.K.), and Geriatric Medicine (H.A.), Kyoto University Graduate School of Medicine (H.A., T.A.), Translational Research Center, Kyoto University Hospital (H.A., T.A.), Japan
| | - Takashi Akamizu
- From the Departments of Clinical Innovative Medicine (Y.T., M.S., T.M., M.Y.), Cardiovascular Medicine (Y.U., K.O., T.K.), Nephrology (H.K.), and Geriatric Medicine (H.A.), Kyoto University Graduate School of Medicine (H.A., T.A.), Translational Research Center, Kyoto University Hospital (H.A., T.A.), Japan
| | - Toru Kita
- From the Departments of Clinical Innovative Medicine (Y.T., M.S., T.M., M.Y.), Cardiovascular Medicine (Y.U., K.O., T.K.), Nephrology (H.K.), and Geriatric Medicine (H.A.), Kyoto University Graduate School of Medicine (H.A., T.A.), Translational Research Center, Kyoto University Hospital (H.A., T.A.), Japan
| | - Masayuki Yokode
- From the Departments of Clinical Innovative Medicine (Y.T., M.S., T.M., M.Y.), Cardiovascular Medicine (Y.U., K.O., T.K.), Nephrology (H.K.), and Geriatric Medicine (H.A.), Kyoto University Graduate School of Medicine (H.A., T.A.), Translational Research Center, Kyoto University Hospital (H.A., T.A.), Japan
| | - Hidenori Arai
- From the Departments of Clinical Innovative Medicine (Y.T., M.S., T.M., M.Y.), Cardiovascular Medicine (Y.U., K.O., T.K.), Nephrology (H.K.), and Geriatric Medicine (H.A.), Kyoto University Graduate School of Medicine (H.A., T.A.), Translational Research Center, Kyoto University Hospital (H.A., T.A.), Japan
| |
Collapse
|
27
|
Kim MP, Wahl LM, Yanek LR, Becker DM, Becker LC. A monocyte chemoattractant protein-1 gene polymorphism is associated with occult ischemia in a high-risk asymptomatic population. Atherosclerosis 2007; 193:366-72. [PMID: 16934270 DOI: 10.1016/j.atherosclerosis.2006.06.029] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2005] [Revised: 04/27/2006] [Accepted: 06/25/2006] [Indexed: 11/23/2022]
Abstract
Monocyte chemoattractant protein-1 (MCP-1) recruits monocytes into atherosclerotic plaques. A single nucleotide polymorphism in the MCP-1 gene promoter (-2578A>G) results in greater production of MCP-1 protein. We examined the association of this polymorphism with occult coronary artery disease (CAD) and its interaction with CAD risk factor burden, as assessed by the Framingham risk score (FRS) for hard events. We genotyped 679 apparently healthy 24-59-year-old siblings (SIBS) of people with premature CAD, tested for occult ischemia with exercise treadmill tests and thallium-201 single photon emission computed tomography, and assessed CAD risk factors to calculate the FRS. Occult ischemia occurred in 18% of SIBS and overall was somewhat more prevalent in those with the G allele (20.6%) compared to those without (15.6%), p=0.095. In SIBS at higher risk (highest quartile of FRS, >or=6.8%), occult ischemia occurred significantly more frequently in those with the G allele (44.4% versus 26.1%, p=0.017), while there was no significant difference in SIBS with lower FRS. After adjusting for individual risk factors included in the FRS, multivariate logistic regression modeling demonstrated that the G allele independently predicted occult ischemia in the entire study population (p=0.014, OR=1.86, 95% CI=1.14-3.04). This study demonstrates for the first time that the MCP-1 gene -2578A>G polymorphism is associated with an excess risk of coronary atherosclerosis in an asymptomatic population and demonstrates an apparent interaction with CAD risk factor burden.
Collapse
Affiliation(s)
- Min P Kim
- Immunopathology Section, National Institute of Dental and Craniofacial Research, USA
| | | | | | | | | |
Collapse
|
28
|
Yamashita T, Kawashima S, Hirase T, Shinohara M, Takaya T, Sasaki N, Takeda M, Tawa H, Inoue N, Hirata KI, Yokoyama M. Xenogenic macrophage immunization reduces atherosclerosis in apolipoprotein E knockout mice. Am J Physiol Cell Physiol 2007; 293:C865-73. [PMID: 17553935 DOI: 10.1152/ajpcell.00117.2007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Atherosclerosis is a complex chronic inflammatory disease in which macrophages play a critical role, and the intervention of the inflammatory process in atherogenesis could be a therapeutic strategy. In this study, we investigated the efficacy of xenogenic macrophage immunization on the atherosclerotic lesion formation in a model of murine atherosclerosis. Apolipoprotein E knockout (apoE-KO) mice were repeatedly immunized with formaldehyde-fixed cultured human macrophages (phorbol ester-stimulated THP-1 cells), using human serum albumin as a control protein or HepG2 cells as human control cells, once a week for four consecutive weeks. The vehicle phosphate-buffered saline was injected in the nonimmunized controls. THP-1 immunization induced antibodies that are immunoreactive with mouse macrophages. Although the plasma lipid levels were unchanged by the immunization, the atherosclerotic lesion area in the aortic root was significantly reduced by >50% in 16-wk-old THP-1-immunized apoE-KO mice compared with that in control mice. THP-1 immunization reduced in vivo macrophage infiltration, reduced in vitro macrophage adhesion, and changed cytokine production by macrophages to the antiatherogenic phenotype. Xenogenic macrophage immunization protects against the development of atherosclerosis in apoE-KO mice by modulating macrophage function in which antibodies induced by the immunization are likely to be involved. This method is a novel and potentially useful cell-mediated immune therapeutic technique against atherosclerosis.
Collapse
Affiliation(s)
- Tomoya Yamashita
- Division of Cardiovascular Medicine, Dept. of Internal Medicine, Kobe Univ. Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
The emigration of leucocytes into the tissue as a crucial step in the response to inflammatory signals has been acknowledged for more than 100 years. The endothelium does not only represent a mechanical barrier between blood and tissue, the circulatory system also connects different organ systems with each other, thus allowing the communication between remote systems. Leukocytes can function as messengers and messages at the same time. Failure or dysregulation of leucocyte-endothelial communication can severely affect the integrity of the organism. The interaction between leucocytes and the vascular endothelium has been recognised as an attractive target for the therapy of numerous disorders and diseases, including excessive inflammatory responses and autoimmune diseases, both associated with enormous consequences for patients and the health care system. There is promising evidence that the success rate of such treatments will increase as the understanding of the molecular mechanisms keeps improving. This chapter reviews the current knowledge about leucocyte-endothelial interaction. It will also display examples of both physiological and dysregulated leucocyte-endothelial interactions and identify potential therapeutical approaches.
Collapse
Affiliation(s)
- K Ley
- Robert M. Berne Cardiovascular Research Center, University of Virginia Health System, 415 Lane Road, MR5 Building, Charlottesville, VA 22903, USA.
| | | |
Collapse
|
30
|
Fan J, Watanabe T. Inflammatory reactions in the pathogenesis of atherosclerosis. J Atheroscler Thromb 2003; 10:63-71. [PMID: 12740479 DOI: 10.5551/jat.10.63] [Citation(s) in RCA: 226] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Atherosclerosis and its complications constitute the most common causes of death in Western societies and Japan. Although several theories or hypotheses about atherogenesis have been proposed during the past decades, none can completely explain the whole process of the pathogenesis of atherosclerosis because this disease is associated with multiple risk factors. In spite of this, the concept that atherosclerosis is a specific form of chronic inflammatory process resulting from interactions between plasma lipoproteins, cellular components ( monocyte/macrophages, T lymphocytes, endothelial cells and smooth muscle cells ) and the extracellular matrix of the arterial wall, is now well accepted. Histologically, atherosclerotic lesions from the early-stage ( fatty streak ) to more complicated lesions possess all the features of chronic inflammation. It has been demonstrated that atherogenic lipoproteins such as oxidized low density lipoprotein ( LDL ), remnant lipoprotein (beta-VLDL) and lipoprotein [ Lp ] ( a ) play a critical role in the pro-inflammatory reaction, whereas high density lipoprotein ( HDL ), anti-atherogenic lipoproteins, exert anti-inflammatory functions. In cholesterol-fed animals, the earliest events in the arterial wall during atherogenesis are the adhesion of monocytes and lymphocytes to endothelial cells followed by the migration of these cells into the intima. It has been shown that these early events in atherosclerosis are triggered by the presence of high levels of atherogenic lipoproteins in the plasma and are mediated by inflammatory factors such as adhesion molecules and cytokines in the arterial wall. The development of genetically modified laboratory animals ( transgenic and knock-out mice and transgenic rabbits ) has provided a powerful approach for dissecting individual candidate genes and studying their cause-and-effect relationships in lesion formation and progression. The purpose of this article is to review the recent progress regarding the inflammatory processes during the development of atherosclerosis based on both human and experimental studies. In particular, we will address the mechanisms of atherogenic lipoproteins in terms of inflammatory reactions associated with hypercholesterolemia. Understanding the molecular mechanisms responsible for inflammatory reactions during atherogenesis may help us to develop novel therapeutic strategies to control, treat and prevent atherosclerosis in the future.
Collapse
Affiliation(s)
- Jianglin Fan
- Laboratory of Cardiovascular Disease, Institute of Basic Medical Sciences, University of Tsukuba, Tsukuba, Japan
| | | |
Collapse
|
31
|
Waksman R, Pakala R, Burnett MS, Gulick CP, Leborgne L, Fournadjiev J, Wolfram R, Hellinga D. Oral rapamycin inhibits growth of atherosclerotic plaque in apoE knock-out mice. CARDIOVASCULAR RADIATION MEDICINE 2003; 4:34-8. [PMID: 12892771 DOI: 10.1016/s1522-1865(03)00121-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Inflammatory and immunological responses of vascular cells are known to play significant roles in atherosclerotic plaque development. Rapamycin with antiinflammatory, immunosuppressive and antiproliferative properties has been shown to reduce neointima formation when coated on stents. This study is designed to test the potential of oral rapamycin to inhibit atherosclerotic plaque development. METHODS Eight-week-old apoE knock-out mice were fed with 0.25% cholesterol supplemented diet (control diet), control diet containing 50 microg/kg rapamycin (low-dose rapamycin) or 100 microg/kg rapamycin (high-dose rapamycin) for 4 or 8 weeks. Subsets of mice from each group (n=10) were weighed and euthanized. Whole blood rapamycin levels were determined using HPLC-MS/MS, and histological analyses of atherosclerotic lesions in the aortic root were performed. RESULTS Mice fed with high-dose rapamycin did not gain weight (18.5+/-1.5 vs. 20.6+/-0.9 g, P=.01). Blood levels of rapamycin 117+/-7 pg/ml were detected in the blood of mice fed with high-dose rapamycin for 8 weeks. The plaque area in mice fed with high dose oral rapamycin is significantly less as compared to control (0.168+/-0.008 vs. 0.326+/-0.013 mm2, P=.001 at 4 weeks; 0.234+/-0.013 vs. 0.447+/-0.011 mm2, P=.001 at 8 weeks). Lumen area was inversely proportional to the plaque area. CONCLUSIONS The results indicate that oral rapamycin is effective in attenuating the progression of atherosclerotic plaque in the mice.
Collapse
Affiliation(s)
- Ron Waksman
- Cardiovascular Research Institute, Washington Hospital Center, 110 Irving Street, NW, Suite 4B-1, Washington, DC 20010, USA.
| | | | | | | | | | | | | | | |
Collapse
|