1
|
Yang H, Li J, Huang XR, Bucala R, Xu A, Lan HY. Macrophage-derived macrophage migration inhibitory factor mediates renal injury in anti-glomerular basement membrane glomerulonephritis. Front Immunol 2024; 15:1361343. [PMID: 38846956 PMCID: PMC11153660 DOI: 10.3389/fimmu.2024.1361343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 04/30/2024] [Indexed: 06/09/2024] Open
Abstract
Macrophages are a rich source of macrophage migration inhibitory factor (MIF). It is well established that macrophages and MIF play a pathogenic role in anti-glomerular basement membrane crescentic glomerulonephritis (anti-GBM CGN). However, whether macrophages mediate anti-GBM CGN via MIF-dependent mechanism remains unexplored, which was investigated in this study by specifically deleting MIF from macrophages in MIFf/f-lysM-cre mice. We found that compared to anti-GBM CGN induced in MIFf/f control mice, conditional ablation of MIF in macrophages significantly suppressed anti-GBM CGN by inhibiting glomerular crescent formation and reducing serum creatinine and proteinuria while improving creatine clearance. Mechanistically, selective MIF depletion in macrophages largely inhibited renal macrophage and T cell recruitment, promoted the polarization of macrophage from M1 towards M2 via the CD74/NF-κB/p38MAPK-dependent mechanism. Unexpectedly, selective depletion of macrophage MIF also significantly promoted Treg while inhibiting Th1 and Th17 immune responses. In summary, MIF produced by macrophages plays a pathogenic role in anti-GBM CGN. Targeting macrophage-derived MIF may represent a novel and promising therapeutic approach for the treatment of immune-mediated kidney diseases.
Collapse
Affiliation(s)
- Hui Yang
- Department of Nephrology, Sun Yat‐Sen Memorial Hospital, Sun Yat‐Sen University, Guangzhou, China
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jinhong Li
- Department of Nephrology, The Seventh Affiliated Hospital of Sun Yat‐sen University, SunYat‐sen University, Shenzhen, China
| | - Xiao-ru Huang
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Departments of Nephrology and Pathology, Guangdong Provincial Hospital, Southern Medical University, Guangzhou, China
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Anping Xu
- Department of Nephrology, Sun Yat‐Sen Memorial Hospital, Sun Yat‐Sen University, Guangzhou, China
| | - Hui-Yao Lan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Departments of Nephrology and Pathology, Guangdong Provincial Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
2
|
Chai L, Wang Q, Wang Y, Li D, Zhang Q, Chen Y, Liu J, Chen H, Qiu Y, Shen N, Wang J, Xie X, Li M. Downregulation of PDCD4 through STAT3/ATF6/autophagy mediates MIF-induced PASMCs proliferation/migration and vascular remodeling. Eur J Pharmacol 2023; 956:175968. [PMID: 37549728 DOI: 10.1016/j.ejphar.2023.175968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 07/08/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
To address the molecular mechanisms underlying macrophage migration inhibitory factor (MIF) induced pulmonary artery smooth muscle cells (PASMCs) proliferation, migration and vascular remodeling in pulmonary hypertension (PH), primary cultured rat PASMCs and monocrotaline (MCT)-induced rats with PH were applied in the present study. The results showed that MIF increased signal transducer and activator of transcription 3 (STAT3) phosphorylation, and then stimulated activating transcription factor 6 (ATF6) activation, subsequently triggered autophagy activation, which further led to programmed cell death factor 4 (PDCD4) lysosomal degradation, and eventually promoted PASMCs proliferation/migration. In lung tissues of MCT rats, MIF protein expression was elevated, phosphorylation of STAT3 and activation of ATF6 were increased, activation of autophagy was evident, and reduction of PDCD4 was observed. Intervention with MIF inhibitor 4-Iodo-6-phenylpyrimidine (4-IPP), ATF6 blocker melatonin or autophagy inhibitor chloroquine, confirmed the in vitro interaction among MIF, STAT3, ATF6, autophagy and PDCD4 in MCT induced rats with PH. Targeting MIF/STAT3/ATF6/autophagy/PDCD4 axis effectively prevented the development of PH by suppressing PASMCs proliferation and vascular remodeling. In conclusions, we demonstrate that MIF activates the STAT3/ATF6/autophagy cascade and then degrades PDCD4 leading to PASMCs proliferation/migration and pulmonary vascular remodeling, suggesting that intervention this axis might have potential value in management of PH.
Collapse
Affiliation(s)
- Limin Chai
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Qingting Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Yan Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Danyang Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Qianqian Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Yuqian Chen
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Jin Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Huan Chen
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Yuanjie Qiu
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Nirui Shen
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Jian Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Xinming Xie
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Manxiang Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China.
| |
Collapse
|
3
|
Krammer C, Yang B, Reichl S, Besson-Girard S, Ji H, Bolini V, Schulte C, Noels H, Schlepckow K, Jocher G, Werner G, Willem M, El Bounkari O, Kapurniotu A, Gokce O, Weber C, Mohanta S, Bernhagen J. Pathways linking aging and atheroprotection in Mif-deficient atherosclerotic mice. FASEB J 2023; 37:e22752. [PMID: 36794636 DOI: 10.1096/fj.202200056r] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 11/30/2022] [Accepted: 12/21/2022] [Indexed: 02/17/2023]
Abstract
Atherosclerosis is a chronic inflammatory condition of our arteries and the main underlying pathology of myocardial infarction and stroke. The pathogenesis is age-dependent, but the links between disease progression, age, and atherogenic cytokines and chemokines are incompletely understood. Here, we studied the chemokine-like inflammatory cytokine macrophage migration inhibitory factor (MIF) in atherogenic Apoe-/- mice across different stages of aging and cholesterol-rich high-fat diet (HFD). MIF promotes atherosclerosis by mediating leukocyte recruitment, lesional inflammation, and suppressing atheroprotective B cells. However, links between MIF and advanced atherosclerosis across aging have not been systematically explored. We compared effects of global Mif-gene deficiency in 30-, 42-, and 48-week-old Apoe-/- mice on HFD for 24, 36, or 42 weeks, respectively, and in 52-week-old mice on a 6-week HFD. Mif-deficient mice exhibited reduced atherosclerotic lesions in the 30/24- and 42/36-week-old groups, but atheroprotection, which in the applied Apoe-/- model was limited to lesions in the brachiocephalic artery and abdominal aorta, was not detected in the 48/42- and 52/6-week-old groups. This suggested that atheroprotection afforded by global Mif-gene deletion differs across aging stages and atherogenic diet duration. To characterize this phenotype and study the underlying mechanisms, we determined immune cells in the periphery and vascular lesions, obtained a multiplex cytokine/chemokine profile, and compared the transcriptome between the age-related phenotypes. We found that Mif deficiency promotes lesional macrophage and T-cell counts in younger but not aged mice, with subgroup analysis pointing toward a role for Trem2+ macrophages. The transcriptomic analysis identified pronounced MIF- and aging-dependent changes in pathways predominantly related to lipid synthesis and metabolism, lipid storage, and brown fat cell differentiation, as well as immunity, and atherosclerosis-relevant enriched genes such as Plin1, Ldlr, Cpne7, or Il34, hinting toward effects on lesional lipids, foamy macrophages, and immune cells. Moreover, Mif-deficient aged mice exhibited a distinct plasma cytokine/chemokine signature consistent with the notion that mediators known to drive inflamm'aging are either not downregulated or even upregulated in Mif-deficient aged mice compared with the corresponding younger ones. Lastly, Mif deficiency favored formation of lymphocyte-rich peri-adventitial leukocyte clusters. While the causative contributions of these mechanistic pillars and their interplay will be subject to future scrutiny, our study suggests that atheroprotection due to global Mif-gene deficiency in atherogenic Apoe-/- mice is reduced upon advanced aging and identifies previously unrecognized cellular and molecular targets that could explain this phenotype shift. These observations enhance our understanding of inflamm'aging and MIF pathways in atherosclerosis and may have implications for translational MIF-directed strategies.
Collapse
Affiliation(s)
- Christine Krammer
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Bishan Yang
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Sabrina Reichl
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Simon Besson-Girard
- Systems Neuroscience Laboratory, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Munich, Germany.,Graduate School of Systemic Neurosciences (GSN), LMU Munich, Planegg-Martinsried, Germany
| | - Hao Ji
- Systems Neuroscience Laboratory, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Munich, Germany
| | - Verena Bolini
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Corinna Schulte
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Rhenish-Westphalian Technical University (RWTH) Aachen University, Aachen, Germany
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Rhenish-Westphalian Technical University (RWTH) Aachen University, Aachen, Germany.,Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Kai Schlepckow
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Georg Jocher
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Georg Werner
- Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Michael Willem
- Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Omar El Bounkari
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Aphrodite Kapurniotu
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technical University of Munich (TUM), Munich, Germany
| | - Ozgun Gokce
- Systems Neuroscience Laboratory, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Christian Weber
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Institute for Cardiovascular Prevention, LMU University Hospital, Ludwig-Maximilians-University (LMU), Munich, Germany.,Munich Heart Alliance, Munich, Germany
| | - Sarajo Mohanta
- Institute for Cardiovascular Prevention, LMU University Hospital, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Jürgen Bernhagen
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University (LMU), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Munich Heart Alliance, Munich, Germany
| |
Collapse
|
4
|
Siques P, Pena E, Brito J, El Alam S. Oxidative Stress, Kinase Activation, and Inflammatory Pathways Involved in Effects on Smooth Muscle Cells During Pulmonary Artery Hypertension Under Hypobaric Hypoxia Exposure. Front Physiol 2021; 12:690341. [PMID: 34434114 PMCID: PMC8381601 DOI: 10.3389/fphys.2021.690341] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 07/16/2021] [Indexed: 12/23/2022] Open
Abstract
High-altitude exposure results in hypobaric hypoxia, which affects organisms by activating several mechanisms at the physiological, cellular, and molecular levels and triggering the development of several pathologies. One such pathology is high-altitude pulmonary hypertension (HAPH), which is initiated through hypoxic pulmonary vasoconstriction to distribute blood to more adequately ventilated areas of the lungs. Importantly, all layers of the pulmonary artery (adventitia, smooth muscle, and endothelium) contribute to or are involved in the development of HAPH. However, the principal action sites of HAPH are pulmonary artery smooth muscle cells (PASMCs), which interact with several extracellular and intracellular molecules and participate in mechanisms leading to proliferation, apoptosis, and fibrosis. This review summarizes the alterations in molecular pathways related to oxidative stress, inflammation, kinase activation, and other processes that occur in PASMCs during pulmonary hypertension under hypobaric hypoxia and proposes updates to pharmacological treatments to mitigate the pathological changes in PASMCs under such conditions. In general, PASMCs exposed to hypobaric hypoxia undergo oxidative stress mediated by Nox4, inflammation mediated by increases in interleukin-6 levels and inflammatory cell infiltration, and activation of the protein kinase ERK1/2, which lead to the proliferation of PASMCs and contribute to the development of hypobaric hypoxia-induced pulmonary hypertension.
Collapse
Affiliation(s)
- Patricia Siques
- Institute of Health Studies, Arturo Prat University, Iquique, Chile
| | - Eduardo Pena
- Institute of Health Studies, Arturo Prat University, Iquique, Chile
| | - Julio Brito
- Institute of Health Studies, Arturo Prat University, Iquique, Chile
| | - Samia El Alam
- Institute of Health Studies, Arturo Prat University, Iquique, Chile
| |
Collapse
|
5
|
Gencer S, Evans BR, van der Vorst EP, Döring Y, Weber C. Inflammatory Chemokines in Atherosclerosis. Cells 2021; 10:cells10020226. [PMID: 33503867 PMCID: PMC7911854 DOI: 10.3390/cells10020226] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/18/2021] [Accepted: 01/22/2021] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis is a long-term, chronic inflammatory disease of the vessel wall leading to the formation of occlusive or rupture-prone lesions in large arteries. Complications of atherosclerosis can become severe and lead to cardiovascular diseases (CVD) with lethal consequences. During the last three decades, chemokines and their receptors earned great attention in the research of atherosclerosis as they play a key role in development and progression of atherosclerotic lesions. They orchestrate activation, recruitment, and infiltration of immune cells and subsequent phenotypic changes, e.g., increased uptake of oxidized low-density lipoprotein (oxLDL) by macrophages, promoting the development of foam cells, a key feature developing plaques. In addition, chemokines and their receptors maintain homing of adaptive immune cells but also drive pro-atherosclerotic leukocyte responses. Recently, specific targeting, e.g., by applying cell specific knock out models have shed new light on their functions in chronic vascular inflammation. This article reviews recent findings on the role of immunomodulatory chemokines in the development of atherosclerosis and their potential for targeting.
Collapse
Affiliation(s)
- Selin Gencer
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, 80336 Munich, Germany; (S.G.); (E.P.C.v.d.V.); (Y.D.)
| | - Bryce R. Evans
- Department of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (B.R.E.)
| | - Emiel P.C. van der Vorst
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, 80336 Munich, Germany; (S.G.); (E.P.C.v.d.V.); (Y.D.)
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 80336 Munich, Germany
- Interdisciplinary Center for Clinical Research (IZKF), Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Yvonne Döring
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, 80336 Munich, Germany; (S.G.); (E.P.C.v.d.V.); (Y.D.)
- Department of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (B.R.E.)
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, 80336 Munich, Germany; (S.G.); (E.P.C.v.d.V.); (Y.D.)
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 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), 80336 Munich, Germany
- Correspondence:
| |
Collapse
|
6
|
Li DY, Zhang JY, Chen QJ, Liu F, Zhao Q, Gao XM, Li XM, Yang YN. MIF -173G/C (rs755622) polymorphism modulates coronary artery disease risk: evidence from a systematic meta-analysis. BMC Cardiovasc Disord 2020; 20:300. [PMID: 32560699 PMCID: PMC7304150 DOI: 10.1186/s12872-020-01564-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 06/02/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Coronary artery disease (CAD) remains one of the major causes of death in humans. Genetic testing may allow early detection and prevention of this disease. This study aimed to investigate the association between the macrophage migration inhibitory factor (MIF) -173G/C (rs755622) polymorphism and susceptibility to CAD based on a meta-analysis. METHODS We searched several databases to identify observational case-control studies investigating the association between the MIF -173G > C (rs755622) polymorphism and CAD risk published before July 30, 2019. Data were analyzed using the STATA software. RESULTS Six studies, comprising a total of 1172 CAD cases and 1564 controls evaluated for MIF polymorphisms, were included. The occurrence of CAD was found to be associated with the C allele of the MIF rs755622 SNP in the total population (C/G, OR = 1.489, 95% CI = 1.223-1.813). Further, MIF -173G/C polymorphism was significantly associated with CAD under the allelic model in the Asian (C/G, OR = 1.775, 95% CI = 1.365-2.309) and Caucasian (C/G, OR = 1.288, 95% CI 1.003-1.654) subgroups. The data showed that the risk of CAD was higher in the population carrying the C allele. CONCLUSIONS We found evidence of associations between MIF -173C/G and CAD susceptibility in the Asian and Caucasian populations.
Collapse
Affiliation(s)
- De-Yang Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.,Department one of coronary heart disease, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Jin-Yu Zhang
- Rehabilitation department, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Qing-Jie Chen
- Department one of coronary heart disease, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Fen Liu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.,Department one of coronary heart disease, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Qian Zhao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.,Department one of coronary heart disease, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Xiao-Ming Gao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xiao-Mei Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.,Department one of coronary heart disease, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Yi-Ning Yang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China. .,Department one of coronary heart disease, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China.
| |
Collapse
|
7
|
Wu DM, Zheng ZH, Wang S, Wen X, Han XR, Wang YJ, Shen M, Fan SH, Zhang ZF, Shan Q, Li MQ, Hu B, Zheng YL, Chen GQ, Lu J. Association between plasma macrophage migration inhibitor factor and deep vein thrombosis in patients with spinal cord injuries. Aging (Albany NY) 2020; 11:2447-2456. [PMID: 31036774 PMCID: PMC6520010 DOI: 10.18632/aging.101935] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 04/23/2019] [Indexed: 12/17/2022]
Abstract
The patients with spinal cord injury (SCI) suffered significantly higher risk of deep vein thrombosis (DVT) than normal population. The aim was to assess the clinical significance of macrophage migration inhibitory factor (MIF) as the risk factor for DVT in acute SCI patients. 207 Chinese patients were enrolled in this study, including thirty-nine (39) patients (18.8 %; 95 %CI: 13.5 %–24.2 %) diagnosed as DVT at the follow-up of 1 month. Nine (9) of the 39 patients (23.1%) were suspected of thrombosis before the screening. The MIF levels in plasma of DVT patients were significantly higher than DVT-free patients. The risks of DVT would be increased by 11 % (OR unadjusted: 1.11; 95% CI, 1.06–1.17, P<0.001) and 8 % (OR adjusted: 1.08; 1.03–1.14, P=0.001), for each additional 1 ng/ml of MIF level. Furthermore, after MIF was combined with established risk factors, area under the receiver operating characteristic curve (standard error) was increased from 0.82(0.035) to 0.85(0.030). The results showed the potential association between the high MIF levels in plasma and elevated DVT risk in SCI patients, which may assist on early intervention.
Collapse
Affiliation(s)
- Dong-Mei Wu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, P.R. China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, P.R. China
| | - Zi-Hui Zheng
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy, School of Medicine and Life Science, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Shan Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, P.R. China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, P.R. China
| | - Xin Wen
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, P.R. China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, P.R. China
| | - Xin-Rui Han
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, P.R. China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, P.R. China
| | - Yong-Jian Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, P.R. China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, P.R. China
| | - Min Shen
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, P.R. China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, P.R. China
| | - Shao-Hua Fan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, P.R. China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, P.R. China
| | - Zi-Feng Zhang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, P.R. China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, P.R. China
| | - Qun Shan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, P.R. China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, P.R. China
| | - Meng-Qiu Li
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, P.R. China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, P.R. China
| | - Bin Hu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, P.R. China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, P.R. China
| | - Yuan-Lin Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, P.R. China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, P.R. China
| | - Gui-Quan Chen
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, P.R. China
| | - Jun Lu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, Xuzhou, P.R. China.,College of Health Sciences, Jiangsu Normal University, Xuzhou, P.R. China
| |
Collapse
|
8
|
Ahmed M, Miller E. Macrophage migration inhibitory factor (MIF) in the development and progression of pulmonary arterial hypertension. Glob Cardiol Sci Pract 2018; 2018:14. [PMID: 30083544 PMCID: PMC6062764 DOI: 10.21542/gcsp.2018.14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 04/18/2018] [Indexed: 02/06/2023] Open
Abstract
Macrophage migration inhibitory factor (MIF) has been described as a pro-inflammatory cytokine and regulator of neuro-endocrine function. It plays an important upstream role in the inflammatory cascade by promoting the release of other inflammatory cytokines such as TNF-alpha and IL-6, ultimately triggering a chronic inflammatory immune response. As lungs can synthesize and release MIF, many studies have investigated the potential role of MIF as a biomarker in assessment of patients with pulmonary arterial hypertension (PAH) and using anti-MIFs as a new therapeutic modality for PAH.
Collapse
Affiliation(s)
- Mohamed Ahmed
- Neonatal-Perinatal Medicine, Pediatrics Department Cohen Children’s Hospital at New York, Northwell Health System
- The Center for Heart and Lung Research, The Feinstein Institute for Medical Research, Manhasset, New York, USA
- School of Medicine, Hofstra University, Hempstead, New York, USA
| | - Edmund Miller
- The Center for Heart and Lung Research, The Feinstein Institute for Medical Research, Manhasset, New York, USA
- School of Medicine, Hofstra University, Hempstead, New York, USA
- The Elmezzi Graduate School of Molecular Medicine, Manhasset, New York, USA
| |
Collapse
|
9
|
Ekşi F, Pehlivan S, Erdoğan MB, Bayram A, Oğuzkan-Balcı S, Yamak B, Pehlivan M. MBL2 and MIF gene polymorphisms in cardiovascular patients with atherosclerotic lesions undergoing heart valve replacement. BIOTECHNOL BIOTEC EQ 2017. [DOI: 10.1080/13102818.2017.1375864] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Fahriye Ekşi
- Department of Medical Microbiology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Sacide Pehlivan
- Department of Medical Biology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Mustafa Bilge Erdoğan
- Clinic of Cardiovascular Surgery, Faculty of Medicine, Bahçesehir University, Istanbul, Turkey
| | - Ayşen Bayram
- Department of Medical Microbiology, Faculty of Medicine, SANKO University, Gaziantep, Turkey
| | - Sibel Oğuzkan-Balcı
- Department of Medical Biology and Genetics, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Birol Yamak
- Clinic of Cardiovascular Surgery, Medical Park Hospital, Gaziantep, Turkey
| | - Mustafa Pehlivan
- Department of Haematology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| |
Collapse
|
10
|
Tilstam PV, Qi D, Leng L, Young L, Bucala R. MIF family cytokines in cardiovascular diseases and prospects for precision-based therapeutics. Expert Opin Ther Targets 2017; 21:671-683. [PMID: 28562118 DOI: 10.1080/14728222.2017.1336227] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine with chemokine-like functions that increasingly is being studied in different aspects of cardiovascular disease. MIF was first identified as a proinflammatory and pro-survival mediator within the immune system, and a second structurally related MIF family member, D-dopachrome tautomerase (a.k.a. MIF-2), was reported recently. Both MIF family members are released by myocardium and modulate the manifestations of cardiovascular disease, specifically in myocardial ischemia. Areas covered: A scientific overview is provided for the involvement of MIF family cytokines in the inflammatory pathogenesis of atherosclerosis, myocardial infarction, and ischemia-reperfusion injury. We summarize findings of experimental, human genetic and clinical studies, and suggest therapeutic opportunities for modulating the activity of MIF family proteins that potentially may be applied in a MIF allele specific manner. Expert opinion: Knowledge of MIF, MIF-2 and their receptor pathways are under active investigation in different types of cardiovascular diseases, and novel therapeutic opportunities are being identified. Clinical translation may be accelerated by accruing experience with MIF-directed therapies currently in human testing in cancer and autoimmunity.
Collapse
Affiliation(s)
- Pathricia V Tilstam
- a Department of Internal Medicine , Yale University School of Medicine , New Haven , CT , USA
| | - Dake Qi
- a Department of Internal Medicine , Yale University School of Medicine , New Haven , CT , USA.,b Department of Biomedical Sciences , Memorial University of Newfoundland , St. John's , Canada
| | - Lin Leng
- a Department of Internal Medicine , Yale University School of Medicine , New Haven , CT , USA
| | - Lawrence Young
- a Department of Internal Medicine , Yale University School of Medicine , New Haven , CT , USA
| | - Richard Bucala
- a Department of Internal Medicine , Yale University School of Medicine , New Haven , CT , USA
| |
Collapse
|
11
|
Fan Y, Zhang J, Chen CY, Xiao YB, Asico LD, Jose PA, Xu JC, Qian GS, Zeng CY. Macrophage migration inhibitory factor triggers vascular smooth muscle cell dedifferentiation by a p68-serum response factor axis. Cardiovasc Res 2017; 113:519-530. [DOI: 10.1093/cvr/cvx025] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 02/01/2017] [Indexed: 11/14/2022] Open
|
12
|
Hibino N, Best CA, Engle A, Ghimbovschi S, Knoblach S, Nath DS, Ishibashi N, Jonas RA. Novel Association of miR-451 with the Incidence of TEVG Stenosis in a Murine Model. Tissue Eng Part A 2015; 22:75-82. [PMID: 26573748 DOI: 10.1089/ten.tea.2014.0664] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The development of a tissue-engineered vascular graft (TEVG) holds great promise for advancing the field of cardiac surgery. Despite the successful translation of this technology, previous reports identify the primary mode of graft failure as stenosis secondary to intimal hyperplasia. MicroRNAs (miRNAs) regulate gene expression by interfering with mRNA function and recent research has suggested miRNA as a potential therapeutic target. The role of miRNAs in TEVGs during neotissue formation is currently unknown. In this study, we investigated if miRNAs regulate the inhibition of graft stenosis. Biodegradable PGA-P(LA/CL) scaffolds were implanted as inferior vena cava interposition grafts in a murine model (n = 14). Mice were sacrificed 14 days following implantation and TEVGs were harvested for histological analysis and miRNA profiling using Affymetrix miRNA arrays. Graft diameters were measured histologically, and the largest grafts (patent group) and smallest grafts (stenosed group) were profiled (n = 4 for each group). Cell population in each graft was analyzed with immunohistochemistry using antismooth muscle actin (SMA) and antimacrophage (F4/80) antibodies. The graft diameter was significantly greater in the patent group (0.63 ± 0.06 mm) than in the stenosed group (0.17 ± 0.06 mm) (p < 0.01). Cell proliferation was significantly greater in the stenosed grafts than in patent grafts (p < 0.01: SMA [187 ± 11 vs. 77 ± 8 cells] vs. p = 0.025: F4/80 [245 ± 23 vs. 187 ± 11 cells]). MiRNA array of 1416 genes showed that in stenosed grafts, mir-451, mir-338, and mir-466 were downregulated and mir-154 was upregulated. Mir-451 exhibited the greatest difference in expression between stenosed and patent grafts by -3.1-fold. Significant negative correlation was found between the expression of mir-451 and cell proliferation (SMA: r = -0.86, p = 0.003; F4/80: r = -0.89, p = 0.001). Our data, along with previous evidence that mir-451 regulates tumor suppressor genes, suggest that downregulation of mir-451 promotes acute proliferation of macrophages and smooth muscle cells, thereby inducing TEVG stenosis. Adequate expression of mir-451 may be critical for improving TEVG patency.
Collapse
Affiliation(s)
- Narutoshi Hibino
- 1 Department of Cardiac Surgery, Johns Hopkins Hospital, Baltimore, Maryland
| | - Cameron A Best
- 2 Tissue Engineering Program and Surgical Research, Nationwide Children's Hospital , Columbus, Ohio
| | - Alyson Engle
- 3 George Washington University School of Medicine and Health Sciences , Washington, District of Columbia
| | - Svetlana Ghimbovschi
- 4 Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences , Washington, District of Columbia.,5 Research Center for Genetic Medicine, Children's National Medical Center , NW Washington, District of Columbia
| | - Susan Knoblach
- 4 Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences , Washington, District of Columbia.,5 Research Center for Genetic Medicine, Children's National Medical Center , NW Washington, District of Columbia
| | - Dilip S Nath
- 6 Department of Cardiovascular Surgery, Children's National Medical Center , NW Washington, District of Columbia
| | - Nobuyuki Ishibashi
- 6 Department of Cardiovascular Surgery, Children's National Medical Center , NW Washington, District of Columbia
| | - Richard A Jonas
- 6 Department of Cardiovascular Surgery, Children's National Medical Center , NW Washington, District of Columbia
| |
Collapse
|
13
|
van der Vorst EPC, Döring Y, Weber C. MIF and CXCL12 in Cardiovascular Diseases: Functional Differences and Similarities. Front Immunol 2015; 6:373. [PMID: 26257740 PMCID: PMC4508925 DOI: 10.3389/fimmu.2015.00373] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 07/07/2015] [Indexed: 12/11/2022] Open
Abstract
Coronary artery disease (CAD) as part of the cardiovascular diseases is a pathology caused by atherosclerosis, a chronic inflammatory disease of the vessel wall characterized by a massive invasion of lipids and inflammatory cells into the inner vessel layer (intima) leading to the formation of atherosclerotic lesions; their constant growth may cause complications such as flow-limiting stenosis and plaque rupture, the latter triggering vessel occlusion through thrombus formation. Pathophysiology of CAD is complex and over the last years many players have entered the picture. One of the latter being chemokines (small 8-12 kDa cytokines) and their receptors, known to orchestrate cell chemotaxis and arrest. Here, we will focus on the chemokine CXCL12, also known as stromal cell-derived factor 1 (SDF-1) and the chemokine-like function chemokine, macrophage migration-inhibitory factor (MIF). Both are ubiquitously expressed and highly conserved proteins and play an important role in cell homeostasis, recruitment, and arrest through binding to their corresponding chemokine receptors CXCR4 (CXCL12 and MIF), ACKR3 (CXCL12), and CXCR2 (MIF). In addition, MIF also binds to the receptor CD44 and the co-receptor CD74. CXCL12 has mostly been studied for its crucial role in the homing of (hematopoietic) progenitor cells in the bone marrow and their mobilization into the periphery. In contrast to CXCL12, MIF is secreted in response to diverse inflammatory stimuli, and has been associated with a clear pro-inflammatory and pro-atherogenic role in multiple studies of patients and animal models. Ongoing research on CXCL12 points at a protective function of this chemokine in atherosclerotic lesion development. This review will focus on the role of CXCL12 and MIF and their differences and similarities in CAD of high risk patients.
Collapse
Affiliation(s)
- Emiel P C van der Vorst
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich , Munich , Germany
| | - Yvonne Döring
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich , Munich , Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich , Munich , Germany ; German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance , Munich , Germany ; Cardiovascular Research Institute Maastricht (CARIM), Maastricht University , Maastricht , Netherlands
| |
Collapse
|
14
|
Cytokines in atherosclerosis: Key players in all stages of disease and promising therapeutic targets. Cytokine Growth Factor Rev 2015; 26:673-85. [PMID: 26005197 PMCID: PMC4671520 DOI: 10.1016/j.cytogfr.2015.04.003] [Citation(s) in RCA: 327] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 04/27/2015] [Indexed: 02/07/2023]
Abstract
Atherosclerosis, a chronic inflammatory disorder of the arteries, is responsible for most deaths in westernized societies with numbers increasing at a marked rate in developing countries. The disease is initiated by the activation of the endothelium by various risk factors leading to chemokine-mediated recruitment of immune cells. The uptake of modified lipoproteins by macrophages along with defective cholesterol efflux gives rise to foam cells associated with the fatty streak in the early phase of the disease. As the disease progresses, complex fibrotic plaques are produced as a result of lysis of foam cells, migration and proliferation of vascular smooth muscle cells and continued inflammatory response. Such plaques are stabilized by the extracellular matrix produced by smooth muscle cells and destabilized by matrix metalloproteinase from macrophages. Rupture of unstable plaques and subsequent thrombosis leads to clinical complications such as myocardial infarction. Cytokines are involved in all stages of atherosclerosis and have a profound influence on the pathogenesis of this disease. This review will describe our current understanding of the roles of different cytokines in atherosclerosis together with therapeutic approaches aimed at manipulating their actions.
Collapse
|
15
|
Chen L, Zhou X, Fan LX, Yao Y, Swenson-Fields KI, Gadjeva M, Wallace DP, Peters DJM, Yu A, Grantham JJ, Li X. Macrophage migration inhibitory factor promotes cyst growth in polycystic kidney disease. J Clin Invest 2015; 125:2399-412. [PMID: 25961459 DOI: 10.1172/jci80467] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 04/06/2015] [Indexed: 12/31/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is characterized by renal cyst formation, inflammation, and fibrosis. Macrophages infiltrate cystic kidneys, but the role of these and other inflammatory factors in disease progression are poorly understood. Here, we identified macrophage migration inhibitory factor (MIF) as an important regulator of cyst growth in ADPKD. MIF was upregulated in cyst-lining epithelial cells in polycystin-1-deficient murine kidneys and accumulated in cyst fluid of human ADPKD kidneys. MIF promoted cystic epithelial cell proliferation by activating ERK, mTOR, and Rb/E2F pathways and by increasing glucose uptake and ATP production, which inhibited AMP-activated protein kinase signaling. MIF also regulated cystic renal epithelial cell apoptosis through p53-dependent signaling. In polycystin-1-deficient mice, MIF was required for recruitment and retention of renal macrophages, which promoted cyst expansion, and Mif deletion or pharmacologic inhibition delayed cyst growth in multiple murine ADPKD models. MIF-dependent macrophage recruitment was associated with upregulation of monocyte chemotactic protein 1 (MCP-1) and inflammatory cytokine TNF-α. TNF-α induced MIF expression, and MIF subsequently exacerbated TNF-α expression in renal epithelial cells, suggesting a positive feedback loop between TNF-α and MIF during cyst development. Our study indicates MIF is a central and upstream regulator of ADPKD pathogenesis and provides a rationale for further exploration of MIF as a therapeutic target for ADPKD.
Collapse
|
16
|
Yüksel A, Bilgir F, Bilgir O, Calan M, Bozkaya G. Increased circulating macrophage migration inhibitory factor levels are associated with coronary artery disease. Clinics (Sao Paulo) 2015; 70:169-72. [PMID: 26017646 PMCID: PMC4449471 DOI: 10.6061/clinics/2015(03)03] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 01/05/2015] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND To evaluate the macrophage migration inhibitory factor and E-selectin levels in patients with acute coronary syndrome. MATERIALS/METHODS We examined the plasma migration inhibitory factor and E-selectin levels in 87 patients who presented with chest pain at our hospital. The patients were classified into two groups according to their cardiac status. Sixty-five patients had acute myocardial infarction, and 22 patients had non-cardiac chest pain (non-coronary disease). We designated the latter group of patients as the control group. The patients who presented with acute myocardial infarction were further divided into two subgroups: ST-elevated myocardial infarction (n = 30) and non-ST elevated myocardial infarction (n = 35). RESULTS We found higher plasma migration inhibitory factor levels in both acute myocardial infarction subgroups than in the control group. However, the E-selectin levels were similar between the acute myocardial infarction and control patients. In addition, we did not find a significant difference in the plasma migration inhibitory factor levels between the ST elevated myocardial infarction and NST-elevated myocardial infarction subgroups. DISCUSSION The circulating concentrations of migration inhibitory factor were significantly increased in acute myocardial infarction patients, whereas the soluble E-selectin levels were similar between acute myocardial infarction patients and control subjects. Our results suggest that migration inhibitory factor may play a role in the atherosclerotic process.
Collapse
Affiliation(s)
- Arif Yüksel
- Department of Internal Medicine, Izmir Bozyaka Training and Research Hospital, Izmir, Turkey
| | - Ferda Bilgir
- Department of Allergy and Immunology, Katip Çelebi University Medical School, Izmir, Turkey
| | - Oktay Bilgir
- Department of Internal Medicine, Izmir Bozyaka Training and Research Hospital, Izmir, Turkey
| | - Mehmet Calan
- Department of Internal Medicine, Izmir Bozyaka Training and Research Hospital, Izmir, Turkey
| | - Giray Bozkaya
- Department of Biochemistry, Izmir Bozyaka Training and Research Hospital, Izmir, Turkey
| |
Collapse
|
17
|
Abstract
Atherosclerosis is an inflammatory disease of the vessel wall characterized by activation of the innate immune system, with macrophages as the main players, as well as the adaptive immune system, characterized by a Th1-dominant immune response. Cytokines play a major role in the initiation and regulation of inflammation. In recent years, many studies have investigated the role of these molecules in experimental models of atherosclerosis. While some cytokines such as TNF or IFNγ clearly had atherogenic effects, others such as IL-10 were found to be atheroprotective. However, studies investigating the different cytokines in experimental atherosclerosis revealed that the cytokine system is complex with both disease stage-dependent and site-specific effects. In this review, we strive to provide an overview of the main cytokines involved in atherosclerosis and to shed light on their individual role during atherogenesis.
Collapse
Affiliation(s)
- Pascal J H Kusters
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Academic Medical Center, L01-146.1, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University (LMU), Munich, Germany.
| |
Collapse
|
18
|
Sherma ND, Borges CR, Trenchevska O, Jarvis JW, Rehder DS, Oran PE, Nelson RW, Nedelkov D. Mass Spectrometric Immunoassay for the qualitative and quantitative analysis of the cytokine Macrophage Migration Inhibitory Factor (MIF). Proteome Sci 2014; 12:52. [PMID: 25328446 PMCID: PMC4201675 DOI: 10.1186/s12953-014-0052-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 10/02/2014] [Indexed: 12/31/2022] Open
Abstract
Background The cytokine MIF (Macrophage Migration Inhibitory Factor) has diverse physiological roles and is present at elevated concentrations in numerous disease states. However, its molecular heterogeneity has not been previously investigated in biological samples. Mass Spectrometric Immunoassay (MSIA) may help elucidate MIF post-translational modifications existing in vivo and provide additional clarity regarding its relationship to diverse pathologies. Results In this work, we have developed and validated a fully quantitative MSIA assay for MIF, and used it in the discovery and quantification of different proteoforms of MIF in serum samples, including cysteinylated and glycated MIF. The MSIA assay had a linear range of 1.56-50 ng/mL, and exhibited good precision, linearity, and recovery characteristics. The new assay was applied to a small cohort of human serum samples, and benchmarked against an MIF ELISA assay. Conclusions The quantitative MIF MSIA assay provides a sensitive, precise and high throughput method to delineate and quantify MIF proteoforms in biological samples.
Collapse
Affiliation(s)
- Nisha D Sherma
- The Biodesign Institute at Arizona State University, Tempe, AZ 85287 USA
| | - Chad R Borges
- The Biodesign Institute at Arizona State University, Tempe, AZ 85287 USA ; Department of Chemistry & Biochemistry at Arizona State University, Tempe, AZ 85287 USA
| | - Olgica Trenchevska
- The Biodesign Institute at Arizona State University, Tempe, AZ 85287 USA
| | - Jason W Jarvis
- The Biodesign Institute at Arizona State University, Tempe, AZ 85287 USA
| | - Douglas S Rehder
- The Biodesign Institute at Arizona State University, Tempe, AZ 85287 USA
| | - Paul E Oran
- The Biodesign Institute at Arizona State University, Tempe, AZ 85287 USA
| | - Randall W Nelson
- The Biodesign Institute at Arizona State University, Tempe, AZ 85287 USA
| | - Dobrin Nedelkov
- The Biodesign Institute at Arizona State University, Tempe, AZ 85287 USA
| |
Collapse
|
19
|
Roubille F, Busseuil D, Shi Y, Nachar W, Mihalache-Avram T, Mecteau M, Gillis MA, Brand G, Théberge-Julien G, Brodeur MR, Kernaleguen AE, Gombos M, Rhéaume E, Tardif JC. The interleukin-1β modulator gevokizumab reduces neointimal proliferation and improves reendothelialization in a rat carotid denudation model. Atherosclerosis 2014; 236:277-85. [DOI: 10.1016/j.atherosclerosis.2014.07.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 06/20/2014] [Accepted: 07/11/2014] [Indexed: 01/11/2023]
|
20
|
Valdés-Alvarado E, Muñoz-Valle JF, Valle Y, Sandoval-Pinto E, García-González IJ, Valdez-Haro A, De la Cruz-Mosso U, Flores-Salinas HE, Padilla-Gutiérrez JR. Association between the -794 (CATT)5-8 MIF gene polymorphism and susceptibility to acute coronary syndrome in a western Mexican population. J Immunol Res 2014; 2014:704854. [PMID: 25105152 PMCID: PMC4106097 DOI: 10.1155/2014/704854] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 05/30/2014] [Accepted: 06/03/2014] [Indexed: 12/19/2022] Open
Abstract
The macrophage migration inhibitory factor (MIF) is related to the progression of atherosclerosis, which, in turn, is a key factor in the development of acute coronary syndrome (ACS). MIF has a CATT short tandem repeat (STR) at position -794 that might be involved in its expression rate. The aim of this study was to investigate the association between the -794 (CATT)5-8 MIF gene polymorphism and susceptibility to ACS in a western Mexican population. This research included 200 ACS patients classified according to the criteria of the American College of Cardiology (ACC) and 200 healthy subjects (HS). The -794 (CATT)5-8 MIF gene polymorphism was analyzed using a conventional polymerase chain reaction (PCR) technique. The 6 allele was the most frequent in both groups (ACS: 54% and HS: 57%). The most common genotypes in ACS patients and HS were 6/7 and 6/6, respectively, and a significant association was found between the 6/7 genotype and susceptibility to ACS (68% versus 47% in ACS and HS, resp., P = 0.03). We conclude that the 6/7 genotype of the MIF -794 (CATT)5-8 polymorphism is associated with susceptibility to ACS in a western Mexican population.
Collapse
Affiliation(s)
- Emmanuel Valdés-Alvarado
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Edificio Q, Primer Piso, Colonia Independencia, 44350 Guadalajara, JAL, Mexico
- Doctorado en Ciencias Biomédicas, Universidad de Guadalajara, Sierra Mojada 950, Colonia Independencia, 44350 Guadalajara, JAL, Mexico
| | - José Francisco Muñoz-Valle
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Edificio Q, Primer Piso, Colonia Independencia, 44350 Guadalajara, JAL, Mexico
| | - Yeminia Valle
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Edificio Q, Primer Piso, Colonia Independencia, 44350 Guadalajara, JAL, Mexico
| | - Elena Sandoval-Pinto
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Edificio Q, Primer Piso, Colonia Independencia, 44350 Guadalajara, JAL, Mexico
- Doctorado en Ciencias Biomédicas, Universidad de Guadalajara, Sierra Mojada 950, Colonia Independencia, 44350 Guadalajara, JAL, Mexico
| | - Ilian Janet García-González
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Edificio Q, Primer Piso, Colonia Independencia, 44350 Guadalajara, JAL, Mexico
- Doctorado en Genética Humana, Universidad de Guadalajara, Sierra Mojada 950, Colonia Independencia,
44350 Guadalajara, JAL, Mexico
| | - Angélica Valdez-Haro
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Edificio Q, Primer Piso, Colonia Independencia, 44350 Guadalajara, JAL, Mexico
- Doctorado en Genética Humana, Universidad de Guadalajara, Sierra Mojada 950, Colonia Independencia,
44350 Guadalajara, JAL, Mexico
| | - Ulises De la Cruz-Mosso
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Edificio Q, Primer Piso, Colonia Independencia, 44350 Guadalajara, JAL, Mexico
- Doctorado en Ciencias Biomédicas, Universidad de Guadalajara, Sierra Mojada 950, Colonia Independencia, 44350 Guadalajara, JAL, Mexico
| | - Héctor Enrique Flores-Salinas
- IMSS, Centro Medico Nacional de Occidente, Belisario Dominguez 1000, Colonia Independencia, 44340 Guadalajara, JAL, Mexico
| | - Jorgé Ramón Padilla-Gutiérrez
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Edificio Q, Primer Piso, Colonia Independencia, 44350 Guadalajara, JAL, Mexico
| |
Collapse
|
21
|
Strüßmann T, Tillmann S, Wirtz T, Bucala R, von Hundelshausen P, Bernhagen J. Platelets are a previously unrecognised source of MIF. Thromb Haemost 2013; 110:1004-13. [PMID: 23846621 DOI: 10.1160/th13-01-0049] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 06/01/2013] [Indexed: 02/07/2023]
Abstract
Macrophage migration inhibitory factor (MIF) is an inflammatory cytokine with chemokine-like functions and a role in atherogenesis. MIF is secreted by various cells including endothelial cells and macrophages. Platelets are another prominent cell type with a role in atherogenesis and are a rich source of atherogenic chemokines. We asked whether platelets express and secrete MIF. In comparison, CXCL12 release was determined. We examined the subcellular localisation of MIF in platelets/megakaryocytes, studied its co-localisation with other platelet-derived mediators and asked whether platelets contain MIF mRNA. Moreover, we probed the functional role of platelet-derived MIF in inflammatory cell recruitment. Using Western blot and ELISA, we demonstrated and quantitated MIF protein in human and mouse platelets. Applying confocal-microscopy, MIF was found to localise in granular-like structures, but did not co-localise with known platelet cytokines. qPCR indicated that platelets contain low levels of MIF mRNA. ELISA measurements from human platelet supernatants showed that, whereas thrombin and collagen triggered the release of MIF and CXCL12, ADP and oxidised LDL promoted CXCL12 but not MIF secretion. Using Transwell assays, we demonstrated that platelet supernatants promoted monocyte chemotaxis and that this was blocked by neutralising MIF antibodies.This is the first report demonstrating MIF secretion from activated platelets, suggesting that platelets are a previously unrecognised source of MIF in inflammatory processes. There are distinct activating stimuli for MIF and CXCL12 secretion. A substantial portion of the chemotactic capacity of stimulated platelet supernatants is contributed by MIF, suggesting a role for platelet-derived MIF in atherogenic cell recruitment.
Collapse
Affiliation(s)
- T Strüßmann
- Prof. Dr. rer. nat. Jürgen Bernhagen, Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Pauwelsstrasse 30, D-52074 Aachen, Germany, Tel.: +49 241 80 88 840/31/41, Fax: +49 241 80 82 427, E-mail: , Web: www.ukaachen.de/sites/lfg/bcmzb
| | | | | | | | | | | |
Collapse
|
22
|
Tillmann S, Bernhagen J, Noels H. Arrest Functions of the MIF Ligand/Receptor Axes in Atherogenesis. Front Immunol 2013; 4:115. [PMID: 23720662 PMCID: PMC3655399 DOI: 10.3389/fimmu.2013.00115] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 04/29/2013] [Indexed: 12/17/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) has been defined as an important chemokine-like function (CLF) chemokine with an essential role in monocyte recruitment and arrest. Adhesion of monocytes to the vessel wall and their transendothelial migration are critical in atherogenesis and many other inflammatory diseases. Chemokines carefully control all steps of the monocyte recruitment process. Those chemokines specialized in controlling arrest are typically immobilized on the endothelial surface, mediating the arrest of rolling monocytes by chemokine receptor-triggered pathways. The chemokine receptor CXCR2 functions as an important arrest receptor on monocytes. An arrest function has been revealed for the bona fide CXCR2 ligands CXCL1 and CXCL8, but genetic studies also suggested that additional arrest chemokines are likely to be involved in atherogenic leukocyte recruitment. While CXCR2 is known to interact with numerous CXC chemokine ligands, the CLF chemokine MIF, which structurally does not belong to the CXC chemokine sub-family, was surprisingly identified as a non-cognate ligand of CXCR2, responsible for critical arrest functions during the atherogenic process. MIF was originally identified as macrophage migration inhibitory factor (this function being eponymous), but is now known as a potent inflammatory cytokine with CLFs including chemotaxis and leukocyte arrest. This review will cover the mechanisms underlying these functions, including MIF’s effects on LFA1 integrin activity and signal transduction, and will discuss the structural similarities between MIF and the bona fide CXCR2 ligand CXCL8 while emphasizing the structural differences. As MIF also interacts with CXCR4, a chemokine receptor implicated in CXCL12-elicited lymphocyte arrest, the arrest potential of the MIF/CXCR4 axis will also be scrutinized as well as the recently identified role of pericyte MIF in attracting leukocytes exiting through venules as part of the pericyte “motility instruction program.”
Collapse
Affiliation(s)
- Sabine Tillmann
- Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University Aachen, Germany
| | | | | |
Collapse
|
23
|
Asare Y, Schmitt M, Bernhagen J. The vascular biology of macrophage migration inhibitory factor (MIF). Expression and effects in inflammation, atherogenesis and angiogenesis. Thromb Haemost 2013; 109:391-8. [PMID: 23329140 DOI: 10.1160/th12-11-0831] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2012] [Accepted: 12/03/2012] [Indexed: 12/18/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine with chemokine-like functions. MIF is a critical mediator of the host immune and inflammatory response. Dysregulated MIF expression has been demonstrated to contribute to various acute and chronic inflammatory conditions as well as cancer development. More recently, MIF has been identified as an important pro-atherogenic factor. Its blockade could even aid plaque regression in advanced atherosclerosis. Promotion of atherogenic leukocyte recruitment processes has been recognised as a major underlying mechanism of MIF in vascular pathology. However, MIF's role in vascular biology is not limited to immune cell recruitment as recent evidence also points to a role for this mediator in neo-angiogenesis / vasculogenesis by endothelial cell activation and endothelial progenitor cell recruitment. On the basis of introducing MIF's chemokine-like functions, the current article focusses on MIF's role in vascular biology and pathology.
Collapse
Affiliation(s)
- Yaw Asare
- Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Pauwelsstrasse 30, D-52074 Aachen, Germany
| | | | | |
Collapse
|
24
|
Khalyfa A, Kheirandish-Gozal L, Capdevila OS, Bhattacharjee R, Gozal D. Macrophage migration inhibitory factor gene polymorphisms and plasma levels in children with obstructive sleep apnea. Pediatr Pulmonol 2012; 47:1001-11. [PMID: 22451332 PMCID: PMC3405200 DOI: 10.1002/ppul.22560] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2011] [Accepted: 11/28/2011] [Indexed: 11/09/2022]
Abstract
INTRODUCTION Obstructive sleep apnea (OSA) is associated with increased risk for cardiovascular and metabolic dysfunction in both adults and children. In adults with OSA, serum levels of macrophage migration inhibitory factor (MIF) are elevated. Therefore, we assessed plasma MIF levels and MIF allelic variant frequencies in children with and without OSA (NOSA). METHODS A total of 614 consecutive children ages 5-8 years were recruited. Children were divided into those with OSA and NOSA based on the apnea-hypopnea index (AHI). In addition to lipid profile, hsCRP, and fasting insulin and glucose levels, plasma MIF levels were assayed using ELISA, and 28 single nucleotide polymorphisms (SNPs) covering the region were genotyped. Linkage disequilibrium and haplotype blocks were analyzed using Haploview version 4.2 software. RESULTS Morning plasma MIF levels were increased in children with OSA. Of the 28 SNPs tested, the frequency of rs10433310 minor allele was significantly decreased in OSA. This SNP was also associated with reduced fasting insulin and hsCRP levels in OSA. The minor allele frequency of all other 27 SNPs was similar in OSA and NOSA groups. CONCLUSIONS Childhood OSA is associated with higher plasma MIF, hsCRP, and fasting insulin levels that promote cardiometabolic risk, and the MIF gene SNP rs10433310 may account for some of the variance in such risk.
Collapse
Affiliation(s)
- Abdelnaby Khalyfa
- Department of Pediatrics, Comer Children's Hospital, The University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | |
Collapse
|
25
|
Merk M, Mitchell RA, Endres S, Bucala R. D-dopachrome tautomerase (D-DT or MIF-2): doubling the MIF cytokine family. Cytokine 2012; 59:10-7. [PMID: 22507380 PMCID: PMC3367028 DOI: 10.1016/j.cyto.2012.03.014] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 03/09/2012] [Accepted: 03/16/2012] [Indexed: 12/12/2022]
Abstract
D-dopachrome tautomerase (D-DT) is a newly described cytokine and a member of the macrophage migration inhibitory factor (MIF) protein superfamily. MIF is a broadly expressed pro-inflammatory cytokine that regulates both the innate and the adaptive immune response. MIF activates the MAP kinase cascade, modulates cell migration, and counter-acts the immunosuppressive effects of glucocorticoids. For many cell types, MIF also acts as an important survival or anti-apoptotic factor. Circulating MIF levels are elevated in the serum in different infectious and autoimmune diseases, and neutralization of the MIF protein via antibodies or small molecule antagonists improves the outcome in numerous animal models of human disease. Recently, a detailed investigation of the biological role of the closely homologous protein D-DT, which is encoded by a gene adjacent to MIF, revealed an overlapping functional spectrum with MIF. The D-DT protein also is present in most tissues and circulates in serum at similar concentrations as MIF. D-DT binds the MIF cell surface receptor complex, CD74/CD44, with high affinity and induces similar cell signaling and effector functions. Furthermore, an analysis of the signaling properties of the two proteins showed that they work cooperatively, and that neutralization of D-DT in vivo significantly decreases inflammation. In this review, we highlight the similarities and differences between MIF and D-DT, which we propose to designate "MIF-2", and discuss the implication of D-DT/MIF-2 expression for MIF-based therapies.
Collapse
Affiliation(s)
- Melanie Merk
- Center of Integrated Protein Science Munich, Division of Clinical Pharmacology, LMU Munich, Germany
| | | | - Stefan Endres
- Center of Integrated Protein Science Munich, Division of Clinical Pharmacology, LMU Munich, Germany
| | - Richard Bucala
- Internal Medicine Yale University School of Medicine, New Haven, CT, 06520
| |
Collapse
|
26
|
Diverse roles of macrophages in atherosclerosis: from inflammatory biology to biomarker discovery. Mediators Inflamm 2012; 2012:693083. [PMID: 22577254 PMCID: PMC3337637 DOI: 10.1155/2012/693083] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2011] [Accepted: 01/11/2012] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease, a leading cause of mortality in developed countries, is mainly caused by atherosclerosis, a chronic inflammatory disease. Macrophages, which differentiate from monocytes that are recruited from the blood, account for the majority of leukocytes in atherosclerotic plaques. Apoptosis and the suppressed clearance of apoptotic macrophages (efferocytosis) are associated with vulnerable plaques that are prone to rupture, leading to thrombosis. Based on the central functions of macrophages in atherogenesis, cytokines, chemokines, enzymes, or microRNAs related to or produced by macrophages have become important clinical prognostic or diagnostic biomarkers. This paper discusses the impact of monocyte-derived macrophages in early atherogenesis and advanced disease. The role and possible future development of macrophage inflammatory biomarkers are also described.
Collapse
|
27
|
Zhang Y, Talwar A, Tsang D, Bruchfeld A, Sadoughi A, Hu M, Omonuwa K, Cheng KF, Al-Abed Y, Miller EJ. Macrophage migration inhibitory factor mediates hypoxia-induced pulmonary hypertension. Mol Med 2012; 18:215-23. [PMID: 22113497 DOI: 10.2119/molmed.2011.00094] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Accepted: 11/15/2011] [Indexed: 12/28/2022] Open
Abstract
Pulmonary hypertension (PH) is a devastating disease leading to progressive hypoxemia, right ventricular failure, and death. Hypoxia can play a pivotal role in PH etiology, inducing pulmonary vessel constriction and remodeling. These events lead to increased pulmonary vessel wall thickness, elevated vascular resistance and right ventricular hypertrophy. The current study examined the association of the inflammatory cytokine macrophage migration inhibitory factor (MIF) with chronic lung disease and its role in the development of hypoxia-induced PH. We found that plasma MIF in patients with primary PH or PH secondary to interstitial lung disease (ILD) was significantly higher than in the control group (P = 0.004 and 0.007, respectively). MIF involvement with hypoxia-induced fibroblast proliferation was examined in both a human cell-line and primary mouse cells from wild-type (mif⁺/⁺) and MIF-knockout (mif⁻/⁻) mice. In vitro, hypoxia-increased MIF mRNA, extracellular MIF protein accumulation and cell proliferation. Inhibition of MIF inflammatory activity reduced hypoxia-induced cell proliferation. However, hypoxia only increased proliferation of mif⁻/⁻ cells when they were supplemented with media from mif⁺/⁺ cells. This growth increase was suppressed by MIF inhibition. In vivo, chronic exposure of mice to a normobaric atmosphere of 10% oxygen increased lung tissue expression of mRNA encoding MIF and accumulation of MIF in plasma. Inhibition of the MIF inflammatory active site, during hypoxic exposure, significantly reduced pulmonary vascular remodeling, cardiac hypertrophy and right ventricular systolic pressure. The data suggest that MIF plays a critical role in hypoxia-induced PH, and its inhibition may be beneficial in preventing the development and progression of the disease.
Collapse
Affiliation(s)
- Yinzhong Zhang
- Centers for Heart and Lung Research, The Feinstein Institute for Medical Research, Manhasset, New York, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Macrophage migration inhibitory factor reduces apoptosis in cerebral arteriovenous malformations. Neurosci Lett 2012; 508:84-8. [DOI: 10.1016/j.neulet.2011.12.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 12/11/2011] [Accepted: 12/14/2011] [Indexed: 01/08/2023]
|
29
|
Role of macrophage migration inhibitory factor in the proliferation of smooth muscle cell in pulmonary hypertension. Mediators Inflamm 2012; 2012:840737. [PMID: 22363104 PMCID: PMC3270469 DOI: 10.1155/2012/840737] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Accepted: 10/11/2011] [Indexed: 11/24/2022] Open
Abstract
Pulmonary hypertension (PH) contributes to the mortality of
patients with lung and heart diseases. However, the underlying
mechanism has not been completely elucidated. Accumulating
evidence suggests that inflammatory response may be involved in
the pathogenesis of PH. Macrophage migration inhibitory factor
(MIF) is a critical upstream inflammatory mediator which promotes
a broad range of pathophysiological processes. The aim of the
study was to investigate the role of MIF in the pulmonary vascular
remodeling of hypoxia-induced PH. We found that MIF mRNA and
protein expression was increased in the lung tissues from hypoxic
pulmonary hypertensive rats. Intensive immunoreactivity for MIF
was observed in smooth muscle cells of large pulmonary arteries
(PAs), endothelial cells of small PAs, and inflammatory cells of
hypoxic lungs. MIF participated in the hypoxia-induced PASMCs
proliferation, and it could directly stimulate proliferation of
these cells. MIF-induced enhanced growth of PASMCs was attenuated
by MEK and JNK inhibitor. Besides, MIF antagonist ISO-1 suppressed
the ERK1/2 and JNK phosphorylation induced by MIF. In conclusion,
the current finding suggested that MIF may act on the
proliferation of PASMCs through the activation of the ERK1/2 and
JNK pathways, which contributes to hypoxic pulmonary hypertension.
Collapse
|
30
|
Macrophage migration inhibitory factor: a multifunctional cytokine in rheumatic diseases. ARTHRITIS 2010; 2010:106202. [PMID: 22046508 PMCID: PMC3195319 DOI: 10.1155/2010/106202] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2010] [Revised: 12/04/2010] [Accepted: 12/21/2010] [Indexed: 01/01/2023]
Abstract
Macrophage migration inhibitory factor (MIF) was originally identified in the culture medium of activated T lymphocytes as a soluble factor that inhibited the random migration of macrophages. MIF is now recognized to be a multipotent cytokine involved in the regulation of immune and inflammatory responses. Moreover, the pivotal nature of its involvement highlights the importance of MIF to the pathogenesis of various inflammatory disorders and suggests that blocking MIF may be a useful therapeutic strategy for treating these diseases. This paper discusses the function and expressional regulation of MIF in several rheumatic diseases and related conditions.
Collapse
|
31
|
Grieb G, Merk M, Bernhagen J, Bucala R. Macrophage migration inhibitory factor (MIF): a promising biomarker. ACTA ACUST UNITED AC 2010; 23:257-64. [PMID: 20520854 DOI: 10.1358/dnp.2010.23.4.1453629] [Citation(s) in RCA: 162] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is an immunoregulatory cytokine, the effect of which on arresting random immune cell movement was recognized several decades ago. Despite its historic name, MIF also has a direct chemokine-like function and promotes cell recruitment. Multiple clinical studies have indicated the utility of MIF as a biomarker for different diseases that have an inflammatory component; these include systemic infections and sepsis, autoimmune diseases, cancer, and metabolic disorders such as type 2 diabetes and obesity. The identification of functional promoter polymorphisms in the MIF gene (MIF) and their association with the susceptibility or severity of different diseases has not only served to validate MIF's role in disease development but also opened the possibility of using MIF genotype information to better predict risk and outcome. In this article, we review the clinical data of MIF and discuss its potential as a biomarker for different disease applications.
Collapse
Affiliation(s)
- Gerrit Grieb
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
| | | | | | | |
Collapse
|
32
|
Greven D, Leng L, Bucala R. Autoimmune diseases: MIF as a therapeutic target. Expert Opin Ther Targets 2010; 14:253-64. [PMID: 20148714 DOI: 10.1517/14728220903551304] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
IMPORTANCE OF THE FIELD Autoimmune inflammatory diseases occur commonly in developed countries. The treatment of these diseases is usually non-curative and is aimed at suppressing inflammatory end-organ damage. Macrophage migration inhibitory factor (MIF) is a multipotent cytokine that has been implicated in the pathogenesis of numerous autoimmune inflammatory disorders. The selective targeting of MIF with either anti-MIF antibody or specific MIF antagonists may offer new therapeutic avenues for these diseases. AREAS COVERED IN THIS REVIEW Our aim is to discuss MIF-directed therapies as a novel therapeutic approach. The review covers literature from the past 10 years. WHAT THE READER WILL GAIN MIF inhibition has been shown to be efficacious in many experimental and pre-clinical studies of autoimmune inflammatory diseases. The close regulatory relationship between MIF and glucocorticoids makes therapeutic antagonism of MIF a potential steroid-sparing therapy in patients with refractory autoimmune diseases. TAKE HOME MESSAGE We expect that MIF antagonism by either small-molecule- or antibody-based approaches will find wide application in the treatment of autoimmune inflammatory diseases. Such therapy also may be informed by the MIF genotype of affected patients.
Collapse
Affiliation(s)
- Dorothee Greven
- Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, Meibergdreef 9, Amsterdam, Netherlands.
| | | | | |
Collapse
|
33
|
Sanchez-Zamora Y, Terrazas LI, Vilches-Flores A, Leal E, Juárez I, Whitacre C, Kithcart A, Pruitt J, Sielecki T, Satoskar AR, Rodriguez-Sosa M. Macrophage migration inhibitory factor is a therapeutic target in treatment of non-insulin-dependent diabetes mellitus. FASEB J 2010; 24:2583-90. [PMID: 20203087 DOI: 10.1096/fj.09-147066] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine involved in the pathogenesis of a variety of autoimmune inflammatory diseases. Here, we investigated the role of MIF in the pathogenesis of non-insulin-dependent diabetes mellitus (NIDDM) using MIF(-/-) mice and a mouse model of streptozotocin (STZ)-induced NIDDM. Following single injection of STZ, MIF(+/+) BALB/c mice showed a significant increase in blood glucose levels, developed polyuria, and succumbed to disease. In contrast, no such increase in blood glucose was observed in MIF(-/-) BALB/c mice treated with STZ. These mice produced significantly less inflammatory cytokines and resistin as compared with MIF(+/+) mice and failed to develop clinical disease. Finally, oral administration of a small-molecule MIF antagonist, CPSI-1306, to outbred ICR mice following induction of NIDDM significantly lowered blood glucose levels in the majority of animals, which was also associated with a significant reduction in the levels of the proinflammatory cytokines IL-6 and TNF-alpha in the sera. Taken together, these results demonstrate that MIF is involved in the pathogenesis of NIDDM and is a therapeutic target to treat this disease.
Collapse
Affiliation(s)
- Yuriko Sanchez-Zamora
- Department of Pathology, Ohio State University Medical Center, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Zernecke A, Weber C. Chemokines in the vascular inflammatory response of atherosclerosis. Cardiovasc Res 2009; 86:192-201. [DOI: 10.1093/cvr/cvp391] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
35
|
Noels H, Bernhagen J, Weber C. Macrophage migration inhibitory factor: a noncanonical chemokine important in atherosclerosis. Trends Cardiovasc Med 2009; 19:76-86. [PMID: 19679264 DOI: 10.1016/j.tcm.2009.05.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
In the recent years, atherogenesis has increasingly been linked to inflammatory processes in the injured vessel wall. Recruitment and arrest of monocytes, T cells, and neutrophils via the concerted actions of multiple chemokines and their chemokine receptors have been the subject of intense research and are being appreciated as key events underlying atherosclerotic lesion formation and progression. The evolutionary conserved cytokine macrophage migration inhibitory factor (MIF) exhibits prominent proinflammatory and proatherogenic functions, and the latest findings on its chemotactic and chemokine-like properties imply MIF as a crucial drug target for the treatment of inflammatory diseases. In this review, the role of MIF in atherosclerosis and injury-induced neointima formation is discussed. We place an emphasis on its proinflammatory and chemokine-like functions in the context of underlying extra- and intracellular signaling mechanisms. These findings clearly distinguish MIF from other cytokines in atherosclerosis and justify the intensive search for inhibitors targeting MIF in the treatment of inflammatory diseases, including advanced atherosclerosis.
Collapse
Affiliation(s)
- Heidi Noels
- Institute of Molecular Cardiovascular Research (IMCAR), 52074 Aachen, Germany
| | | | | |
Collapse
|
36
|
McLean LR, Zhang Y, Li H, Li Z, Lukasczyk U, Choi YM, Han Z, Prisco J, Fordham J, Tsay JT, Reiling S, Vaz RJ, Li Y. Discovery of covalent inhibitors for MIF tautomerase via cocrystal structures with phantom hits from virtual screening. Bioorg Med Chem Lett 2009; 19:6717-20. [PMID: 19836948 DOI: 10.1016/j.bmcl.2009.09.106] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2009] [Revised: 09/25/2009] [Accepted: 09/29/2009] [Indexed: 10/20/2022]
Abstract
Biochemical and X-ray crystallographic studies confirmed that hydroxyquinoline derivatives identified by virtual screening were actually covalent inhibitors of the MIF tautomerase. Adducts were formed by N-alkylation of the Pro-1 at the catalytic site with a loss of an amino group of the inhibitor.
Collapse
Affiliation(s)
- Larry R McLean
- Discovery Research, sanofi-aventis, Bridgewater, NJ 08807, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Chen L, Yang G, Zhang X, Wu J, Gu Q, Wei M, Yang J, Zhu Y, Wang N, Guan Y. Induction of MIF expression by oxidized LDL via activation of NF-kappaB in vascular smooth muscle cells. Atherosclerosis 2009; 207:428-33. [PMID: 19640536 DOI: 10.1016/j.atherosclerosis.2009.05.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2009] [Revised: 05/17/2009] [Accepted: 05/18/2009] [Indexed: 11/25/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine playing important roles in atherosclerosis. MIF gene deficiency and neutralizing antibodies against MIF have been reported to exert anti-atherosclerotic effects in various animal models. However, the mechanism by which MIF is induced in atherosclerotic lesions remains unclear. In the present studies, we cloned a 540bp full-length rabbit MIF cDNA by screening a rabbit uterine library. The cDNA contains a 348bp open-reading frame which encodes a deduced 115-amino acid polypeptide with approximately 90% similarity to human and mouse homologs. Constitutive MIF mRNA expression was detected in most rabbit tissues including aortas. The expression of MIF obviously abounded in vascular smooth muscle cells (VSMCs) of the atherosclerotic plaques. In cultured VSMCs, MIF expression was significantly induced by a pro-atherogenic factor, oxidized low-density lipoprotein (oxLDL). Promoter analysis showed there were two NF-kappaB binding sites in the MIF proximal promoter region. Deletion or mutation of the two sites abolished oxLDL-enhanced MIF promoter activity. Moreover, the induction of MIF by oxLDL can be blocked by IkappaB-alpha overexpression. Taken together, our results revealed that MIF expression can be induced by oxLDL in VSMCs via a NF-kappaB dependent manner, which may contribute to the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Lihong Chen
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Diabetes mellitus and apoptosis: inflammatory cells. Apoptosis 2009; 14:1435-50. [PMID: 19360474 DOI: 10.1007/s10495-009-0340-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Accepted: 03/17/2009] [Indexed: 12/24/2022]
Abstract
Since the early observation that similarities between thyroiditis and insulitis existed, the important role played by inflammation in the development of diabetes has been appreciated. More recently, experiments have shown that inflammation also plays a prominent role in the development of target organ damage arising as complications, with both elements of the innate and the adaptive immune system being involved, and that cytokines contributing to local tissue damage may arise from both infiltrating and resident cells. This review will discuss the experimental evidence that shows that inflammatory cell-mediated apoptosis contributes to target organ damage, from beta cell destruction to both micro- and macro-vascular disease complications, and also how alterations in leukocyte turnover affects immune function.
Collapse
|
39
|
Tereshchenko IP, Petrkova J, Mrazek F, Lukl J, Maksimov VN, Romaschenko AG, Voevoda MI, Petrek M. The macrophage migration inhibitory factor (MIF) gene polymorphism in Czech and Russian patients with myocardial infarction. Clin Chim Acta 2009; 402:199-202. [PMID: 19167373 DOI: 10.1016/j.cca.2008.12.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2008] [Revised: 12/23/2008] [Accepted: 12/24/2008] [Indexed: 10/21/2022]
Abstract
BACKGROUND Macrophage migration inhibitory factor (MIF) is a cytokine implicated in early and advanced atherosclerosis. The aim of this study was to investigate whether polymorphism of MIF gene is associated with myocardial infarction (MI). METHODS Single nucleotide polymorphism (SNP) in MIF gene (-173G/C, rs755622) was investigated in Czech (n=219) and Russian (n=240) MI patients and population control from the same geographical areas (Czech, n=137; Russian, n=174). Further, another SNP (rs1007888) located within the 3' flanking region of the MIF gene was investigated in Czech MI patients and control subjects. RESULTS There were no significant differences in the distribution of MIF -173G/C genotypes, alleles or carriage rates between case and control groups in either populations. However, the GG genotype of the MIF SNP rs1007888 was associated with MI in Czech female patients (p=0.027). CONCLUSIONS Taken together with previous reports, our study suggests that particular MIF gene polymorphisms may contribute to MI susceptibility in females.
Collapse
|
40
|
Proteomic profiling in early venous stenosis formation in a porcine model of hemodialysis graft. J Vasc Interv Radiol 2008; 20:241-51. [PMID: 19028119 DOI: 10.1016/j.jvir.2008.10.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2008] [Revised: 10/01/2008] [Accepted: 10/02/2008] [Indexed: 01/15/2023] Open
Abstract
PURPOSE To use proteomic analysis to identify up- and downregulated proteins in early venous stenosis formation in a porcine model of hemodialysis graft failure. MATERIALS AND METHODS Pigs had chronic renal insufficiency created by subtotal renal infarction caused by renal artery embolization. Arteriovenous polytetrafluoroethylene grafts were placed 28 days later and the animals were killed after a further 3 days (n = 4), 7 days (n = 4), or 14 days (n = 4). Proteomic analysis with isotope-coded affinity tags and multidimensional liquid chromatography followed by tandem mass spectrometry was performed on the venous stenosis and control vessels. Expression of proteins was further confirmed by Western blot analysis. The blood urea nitrogen (BUN) and creatinine levels were determined before renal artery embolization and at the time of graft placement. RESULTS At graft placement, mean BUN and creatinine levels were significantly higher than before embolization (P < .05). Six proteins were identified that were common to all four animals at the same time point. Five proteins (alpha-fetoprotein, fetuin A, macrophage migration inhibitory factor, pyruvate dehydrogenase E1 component, and lactoferrin) were upregulated and one protein (decorin) was downregulated. Expression of macrophage migration inhibitory factor, alpha-fetoprotein, and lactoferrin was further validated with Western blotting. By day 14, lactoferrin and fetuin-A expression were increased significantly in early venous stenosis formation. CONCLUSIONS Significantly increased expression of lactoferrin and fetuin-A were observed in early venous stenosis by day 14. Understanding the role of lactoferrin and fetuin-A in hemodialysis vascular access failure could help in improving outcomes in patients undergoing hemodialysis.
Collapse
|
41
|
Abstract
The highly conserved and archetypical yet atypical cytokine macrophage migration inhibitory factor (MIF) fulfills pleiotropic immune functions in many acute and chronic inflammatory diseases. Recent evidence has emerged from both expression and functional studies to implicate MIF in various aspects of cardiovascular disease. The present review is aimed at providing a synopsis of the involvement of MIF in the inflammatory pathogenesis of atherosclerosis and its consequences, namely unstable plaque formation, remodeling after arterial injury, aneurysm formation, myocardial infarction, or ischemia-reperfusion injury. In addition, other forms of myocardial dysfunction and inflammation and the role of MIF in angiogenesis are reviewed. The functional data are reconciled with recent progress in the identification of heptahelical (CXC chemokine) receptors for MIF, its prototypic role as their noncanonical ligand, and its signal transduction profile operative in atherogenic and inflammatory recruitment of mononuclear cells and in the oxidative damage and apoptosis of cardiomyocytes. Its unique features and functions clearly distinguish MIF from other cytokines implicated in atherogenesis and make it a prime target for achieving therapeutic regression of atherosclerosis. The potential of targeting or exploiting MIF for therapeutic strategies or as a diagnostic marker in the management of cardiovascular diseases or disorders is scrutinized.
Collapse
Affiliation(s)
- Alma Zernecke
- Institute for Molecular Cardiovascular Research, Molekulare Herz-Kreislaufforschung, RWTH Aachen University, Aachen, Germany
| | | | | |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW The present review is to summarize recent advances in molecular mechanisms that regulate vascular smooth muscle cell apoptosis during vascular remodeling. In normal blood vessels apoptosis counteracts cell division, whereas apoptosis is especially crucial for regulating vascular remodeling during cardiovascular diseases. RECENT FINDINGS Recent results have expanded our knowledge regarding the signaling pathways and molecules that regulate vascular smooth muscle cell death in postnatal vascular remodeling. Compelling data from genetic disorders associated with vascular smooth muscle cell loss (e.g., Hutchinson-Gilford progeria syndrome) and experimental studies suggest that changes in hemodynamic and mechanical forces are major modulators for vascular smooth muscle cell apoptosis. Furthermore, understanding the therapeutic effects of antihypertensive drugs related to apoptosis may identify pathways that can improve outcomes independent of the blood pressure fall. SUMMARY Regulation of vascular smooth muscle cell apoptosis is a potential target to modify pathological vascular remodeling and new drugs development.
Collapse
|
43
|
Kleemann R, Zadelaar S, Kooistra T. Cytokines and atherosclerosis: a comprehensive review of studies in mice. Cardiovasc Res 2008; 79:360-76. [PMID: 18487233 PMCID: PMC2492729 DOI: 10.1093/cvr/cvn120] [Citation(s) in RCA: 460] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In the past few years, inflammation has emerged as a major driving force of atherosclerotic lesion development. It is now well-established that from early lesion to vulnerable plaque formation, numerous cellular and molecular inflammatory components participate in the disease process. The most prominent cells that invade in evolving lesions are monocyte-derived macrophages and T-lymphocytes. Both cell types produce a wide array of soluble inflammatory mediators (cytokines, chemokines) which are critically important in the initiation and perpetuation of the disease. This review summarizes the currently available information from mouse studies on the contribution of a specified group of cytokines expressed in atherosclerotic lesions, viz. interleukins (IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-10, IL-12, IL-18, IL-20) and macrophage-associated cytokines [tumour necrosis factor-α (TNF-α); macrophage migration inhibitory factor (MIF); interferon-γ (IFN-γ); colony stimulating factors G-CSF,-M-CSF,-GM-CSF) to atherogenesis. Emphasis is put on the consistency of the effects of these cytokines, i.e. inasmuch an effect depends on the experimental approach applied (overexpression/deletion, strain, gender, dietary conditions, and disease stage). An important outcome of this survey is (i) that only for a few cytokines there is sufficient consistent data allowing classifying them as typically proatherogenic (IL-1, IL-12, IL-18, MIF, IFN-γ, TNF-α, and M-CSF) or antiatherogenic (IL-10) and (ii) that some cytokines (IL-4, IL-6 and GM-CSF) can exert pro- or anti-atherogenic effects depending on the experimental conditions. This knowledge can be used for improved early detection, prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Robert Kleemann
- TNO-BioSciences, Gaubius-Laboratory, Department of Vascular and Metabolic Diseases, PO Box 2215, 2301 CE Leiden, The Netherlands.
| | | | | |
Collapse
|
44
|
Expression of hypoxia inducible factor-1 alpha, macrophage migration inhibition factor, matrix metalloproteinase-2 and -9, and their inhibitors in hemodialysis grafts and arteriovenous fistulas. J Vasc Interv Radiol 2008; 19:252-9. [PMID: 18341958 DOI: 10.1016/j.jvir.2007.10.031] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2007] [Revised: 10/14/2007] [Accepted: 10/15/2007] [Indexed: 12/27/2022] Open
Abstract
PURPOSE It is well recognized that arteriovenous fistulas (AVFs) used for hemodialysis access have better primary patency rates with less restenosis than polytetrafluoroethylene (PTFE) grafts; however, the mechanism responsible for this is not known. Recent data suggest that hypoxia inducible factor-1 alpha (HIF-1 alpha) is associated with vascular restenosis, possibly through mechanisms that increase the production of macrophage migration inhibition factor (MIF), matrix metalloproteinase-2 (MMP-2) and MMP-9, and their inhibitors (tissue inhibitor of MMPs; TIMP). The present study tested the hypothesis that there are differences in the expression patterns of HIF-1 alpha, MIF, MMP-2, MMP-9, and TIMPs in specimens removed from patients with AVFs and PTFE grafts. MATERIALS AND METHODS Whole-vessel tissue samples were obtained from the vein distal to the vein-to-PTFE graft anastomosis and the proximal outflow vein (within 6 cm of the arteriovenous anastomosis) of AVFs from 17 patients who required a surgical revision for thrombosis and stenosis. Nonstenotic veins of four patients undergoing hemodialysis vascular access placement were used as controls. PTFE grafts (n = 6), AVFs (n = 6), and control samples (n = 3) underwent Western blot analysis and zymography. A separate group of five patients with PTFE hemodialysis grafts and one control subject were used for immunohistochemical analysis. RESULTS Specimens from patients with PTFE grafts had significantly higher expression of HIF-1 alpha (P = .03), MIF (P = .02), TIMP-1 (P = .0006), pro-MMP-2 (P = .02), and pro-MMP-9 (P = .046) compared with control veins. The expression of only pro-MMP-9 was significantly higher in AVFs compared with control samples (P = .004). There was a significant increase in the expression of MIF (P = .007) and TIMP-1 (P < .0001) in PTFE graft specimens compared with AVFs. MIF and TIMP-1 were localized to the adventitia of the vein distal to the vein-to-PTFE graft anastomosis. CONCLUSIONS There were major differences in the expression patterns of hypoxia (ie, HIF-1 alpha) and proteins regulated by HIF-1?, including MIF, pro-MMP-2, pro-MMP-9, and TIMP-1, in specimens removed from patients with PTFE grafts and AVFs. Understanding the role of HIF-1 alpha and these proteins in hemodialysis access failure can help improve outcomes.
Collapse
|
45
|
Schober A, Bernhagen J, Weber C. Chemokine-like functions of MIF in atherosclerosis. J Mol Med (Berl) 2008; 86:761-70. [PMID: 18385967 DOI: 10.1007/s00109-008-0334-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2008] [Revised: 02/18/2008] [Accepted: 02/22/2008] [Indexed: 12/22/2022]
Abstract
The cytokine macrophage migration inhibitory factor (MIF) is a unique pro-inflammatory regulator of many acute and chronic inflammatory diseases. In the pathogenesis of atherosclerosis, chronic inflammation of the arterial wall characterized by chemokine-mediated influx of leukocytes plays a central role. The contribution of MIF to atherosclerotic vascular disease has come into focus of many studies in recent years. MIF is highly expressed in macrophages and endothelial cells of different types of atherosclerotic plaques, and functional studies established the contribution of MIF to lesion progression and plaque inflammation. This proatherogenic effect may partly be explained by the finding that MIF regulates inflammatory cell recruitment to lesion areas. Similar to chemokines, MIF induces integrin-dependent arrest and transmigration of monocytes and T cells. These chemokine-like functions are mediated through interaction of MIF with the chemokine receptors CXCR2 and CXCR4 as a non-canonical ligand. In atherogenic monocyte recruitment, MIF-induced monocyte adhesion involves CD74 and CXCR2, which form a signaling receptor complex. In addition to lesion progression, MIF has been implicated in plaque destabilization, since MIF is predominantly expressed in vulnerable plaques and can induce collagen-degrading matrix metalloproteinases. The latter could be a relevant mechanism in atherosclerotic abdominal aneurysm formation, where MIF expression is correlated with aneurysmal expansion. In summary, MIF has been identified as an important regulator of atherosclerotic vascular disease with exceptional chemokine-like functions. Detailed analysis of the interaction of MIF with its receptors could provide valuable information for drug development for the anti-inflammatory treatment of established and unstable atherosclerosis.
Collapse
Affiliation(s)
- Andreas Schober
- Cardiology Unit, Medical Policlinic-City Center Campus, University of Munich, Munich, Germany
| | | | | |
Collapse
|
46
|
Okamoto T, Atsumi T, Shimizu C, Yoshioka N, Koike T. The potential role of macrophage migration inhibitory factor on the migration of vascular smooth muscle cells. J Atheroscler Thromb 2008; 15:13-9. [PMID: 18270460 DOI: 10.5551/jat.e495] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
AIM Macrophage migration inhibitory factor (MIF) is known as a pro-inflammatory cytokine that regulates a broad spectrum of inflammatory reactions. MIF is expressed in vascular smooth muscle cells (VSMCs), and inhibition of the progression of atherosclerosis was observed in MIF-deficient atherosclerotic mice. However, the functional role of MIF in VSMCs has not been elucidated. The aim of this study was to investigate the role of MIF on the migration of VSMCs. METHODS Cultured rat A10 cells, derived from rat embryonic aortic smooth muscle cells, were stimulated with oxLDL, and the effect of MIF knockdown on oxLDL-mediated migration of A10 cells was analyzed. RESULTS Intracellular MIF content was significantly increased and a marked increase of MIF concent-ration was observed in the supernatant of A10 cells treated with oxLDL. The migration of A10 cells was significantly accelerated by the stimulation of recombinant MIF in a dose-dependent manner. Notably, knockdown of intracellular MIF by siRNA abolished oxLDL-induced migration of A10 cells. CONCLUSION These findings suggest that MIF acts on the migration of VSMCs in an autocrine and paracrine fashion. MIF appears to be a novel target for the prevention of cardiovascular events.
Collapse
Affiliation(s)
- Toshiya Okamoto
- Department of Medicine II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | | | | | | | | |
Collapse
|
47
|
Herder C, Illig T, Baumert J, Müller M, Klopp N, Khuseyinova N, Meisinger C, Martin S, Thorand B, Koenig W. Macrophage migration inhibitory factor (MIF) and risk for coronary heart disease: results from the MONICA/KORA Augsburg case-cohort study, 1984-2002. Atherosclerosis 2008; 200:380-8. [PMID: 18242614 DOI: 10.1016/j.atherosclerosis.2007.12.025] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2007] [Revised: 12/07/2007] [Accepted: 12/18/2007] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Macrophage migration inhibitory factor (MIF), a central cytokine of the innate immunity, has been reported to contribute to the development of cardiovascular disease. MIF is expressed in atherosclerotic lesions in humans, and gene deletion and antibody inhibition studies in animal models indicated that MIF may be cause rather than consequence of atherosclerosis. We sought to assess the triangular association between MIF genotypes, circulating MIF levels and risk for incident coronary heart disease (CHD) in the large, prospective, population-based MONICA/KORA case-cohort study (Augsburg, Southern Germany). METHODS MIF genotypes, haplotypes and serum concentrations were determined in 363 individuals with incident CHD and 1908 individuals without CHD during follow-up (mean follow-up time 10.3 years). RESULTS Circulating MIF concentrations were not associated with the risk for CHD. In women, carriers of the minor alleles rs755622C and rs2070766G had a higher risk for incident CHD, and a haplotype that contained these two minor alleles was significantly associated with increased risk for CHD (HR 2.44, 95%CI 1.30-4.59). CONCLUSION The lack of association between serum levels and incident CHD indicates that MIF may not be a novel biomarker for CHD risk. However, the association of a haplotype containing the rs755622C allele, which has been reported before to increase the susceptibility for various other proinflammatory conditions, with CHD points towards a role for MIF in local vascular inflammation and atherogenesis.
Collapse
Affiliation(s)
- Christian Herder
- Institute for Clinical Diabetes Research, German Diabetes Center, Leibniz Institute at Heinrich Heine University, Düsseldorf, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Tiwari R, Singh V, Barthwal M. Macrophages: An elusive yet emerging therapeutic target of atherosclerosis. Med Res Rev 2008; 28:483-544. [DOI: 10.1002/med.20118] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
49
|
Kwaijtaal M, van der Ven AJ, van Diest R, Bruggeman CA, Bär FWHM, Calandra T, Appels A, Sweep FCGJ. Exhaustion is associated with low macrophage migration inhibitory factor expression in patients with coronary artery disease. Psychosom Med 2007; 69:68-73. [PMID: 17244850 DOI: 10.1097/psy.0b013e31802b8750] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE Macrophage migration inhibitory factor (MIF), a protein secreted by immune cells and the pituitary gland, may be associated with coronary artery disease (CAD) and the mental state of coronary patients. The first origin of MIF suggests positive, the second negative associations. The aim of this study was to explore the direction of the association of MIF with CAD and of MIF with exhaustion, if any. METHODS Participants were 194 patients who had been recently treated by percutaneous coronary intervention (PCI) and who were exhausted at the start of the study. Half entered a behavioral intervention program. MIF, C-reactive protein, interleukin (IL)-6, IL-1 receptor antagonist, and neopterin were measured in blood collected 6 weeks after PCI (baseline) and 6 and 18 months after baseline. A single measurement of MIF was also available for 129 age- and sex-matched healthy individuals (reference group). RESULTS At baseline, MIF in patients undergoing PCI was significantly lower than in the reference group (p < .01). New cardiac events occurred twice as often in the lowest quartile than in the highest quartile of MIF concentrations. However, the association was not significant (chi(2) = 2.27; df = 3; p = .52). During follow up, MIF concentrations increased significantly in patients undergoing PCI (p < .001). At 18 months, MIF concentrations were significantly lower in the exhausted patients than in the nonexhausted patients (p = .02). hsCRP, IL-1ra, IL-6, and neopter in concentrations did not change over this time period. CONCLUSIONS The data are suggestive of a negative association of MIF with CAD and of MIF with exhaustion. The observation that those patients who remained exhausted had lower concentrations of MIF fits into earlier observations that suggested that exhausted coronary patients may be characterized by a hypoactivity of the hypothalamic-pituitary-adrenocortical axis.
Collapse
Affiliation(s)
- Martijn Kwaijtaal
- Department of Medical Microbiology, University Hospital Maastricht, P. Debeyeplein 25, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Atherosclerosis as the underlying mechanisms of myocardial infarction, stroke and peripheral artery disease remains the major cause of morbidity and mortality in developed countries. Recent developments in vascular biology have indicated that atherosclerosis can be best characterized as a chronic inflammatory disease of the vessel wall that promotes lesion development and progression. Chemokines regulate and control these processes by orchestrating adhesive interactions of circulating blood cells with the arterial wall and their subsequent extravasation. Exhibiting a high degree of specialization and cooperation, different chemokines mediate distinct steps during the atherogenic recruitment of monocytes and T cells. This diversity of chemokine expression and function might lead to the identification of selective therapeutic targets for the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Elisa A Liehn
- Institute of Molecular Cardiovascular Research (IMCAR), University Hospital, RWTH Aachen, Germany.
| | | | | | | |
Collapse
|