1
|
Pressley KR, Naseem Y, Nalawade S, Forsthuber TG. The distinct functions of MIF in inflammatory cardiomyopathy. Front Immunol 2025; 16:1544484. [PMID: 40092999 PMCID: PMC11906721 DOI: 10.3389/fimmu.2025.1544484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/10/2025] [Indexed: 03/19/2025] Open
Abstract
The immune system plays a crucial role in cardiac homeostasis and disease, and the innate and adaptive immune systems can be beneficial or detrimental in cardiac injury. The pleiotropic proinflammatory cytokine macrophage migration inhibitory factor (MIF) is involved in the pathogenesis of many human disease conditions, including heart diseases and inflammatory cardiomyopathies. Inflammatory cardiomyopathies are frequently observed after microbial infection but can also be caused by systemic immune-mediated diseases, drugs, and toxic substances. Immune cells and MIF are implicated in many of these conditions and may affect progression of inflammatory cardiomyopathy (ICM) to myocardial remodeling and dilated cardiomyopathy (DCM). The potential for targeting MIF therapeutically in patients with inflammatory diseases is an active area of investigation. Here we review the current literature supporting the role(s) of MIF in ICM and cardiac dysfunction. We posit that future research to further elucidate the underlying functions of MIF in cardiac pathologies is warranted.
Collapse
Affiliation(s)
- Kyle R. Pressley
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX, United States
- Department of Neuroscience, Developmental and Regenerative Biology, University of Texas at San Antonio, San Antonio, TX, United States
| | - Yashfa Naseem
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX, United States
| | - Saisha Nalawade
- Department of Pre-clinical Immunology, Corner Therapeutics, Watertown, MA, United States
| | - Thomas G. Forsthuber
- Department of Molecular Microbiology and Immunology, University of Texas at San Antonio, San Antonio, TX, United States
| |
Collapse
|
2
|
Elevated plasma macrophage migration inhibitor factor is associated with hypertension and hypertensive left ventricular hypertrophy. J Hum Hypertens 2023; 37:68-73. [PMID: 35027653 DOI: 10.1038/s41371-022-00657-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 12/21/2021] [Accepted: 01/06/2022] [Indexed: 01/31/2023]
Abstract
Previous studies have found that the macrophage migration inhibitor factor is associated with endothelial dysfunction and ventricular remodelling. The aim of this study was to explore the potential relationship between plasma macrophage migration inhibitor factor levels and hypertension and hypertensive left ventricular hypertrophy. A total of 308 participants (including 187 uncomplicated hypertensive patients and 121 healthy controls) were enroled from 2017 to 2019. The association between macrophage migration inhibitor factors and hypertension and hypertensive left ventricular hypertrophy was estimated with univariate and multivariate logistic regression models. Elevated macrophage migration inhibitor factor was associated with the development of hypertension (second tertile: adjusted OR, 2.27, 95% CI, 1.24-4.16, P = 0.008; third tertile: adjusted OR, 5.43, 95% CI, 2.75-10.71, P < 0.001; compared with the first tertile). In addition, we assessed the association between macrophage migration inhibitor factor and left ventricular hypertrophy in hypertensive patients (n = 187). Plasma macrophage migration inhibitor factor was significantly correlated with hypertensive left ventricular mass index (r = 0.580, P < 0.001). In patients with hypertension, an elevated macrophage migration inhibitor factor was significantly associated with hypertensive left ventricular hypertrophy (second tertile: adjusted OR, 3.20, 95% CI, 1.17-8.78, P = 0.024; third tertile: adjusted OR, 24.95, 95% CI, 8.72-71.41, P < 0.001; compared with the first tertile). Receiver operating characteristic analysis indicated that macrophage migration inhibitor factor had reasonable predictive accuracy for the development of hypertensive left ventricular hypertrophy (area under curve 0.84, 95% CI 0.78-0.90, P < 0.001). Our data indicated that elevated macrophage migration inhibitor factor is associated with hypertension and hypertensive left ventricular hypertrophy.
Collapse
|
3
|
Vyshnevska IR, Storozhenko T, Kopytsya MP, Bila NV, Kis A, Kaaki M. THE ROLE OF BIOMARKER MACROPHAGE MIGRATION INHIBITORY FACTOR IN CARDIAC REMODELING PREDICTION IN PATIENTS WITH ST-SEGMENT ELEVATION MYOCARDIAL INFARCTION. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2023; 76:911-919. [PMID: 37326070 DOI: 10.36740/wlek202305104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
OBJECTIVE The aim: To estimate the role of macrophage migration inhibitory factor and soluble ST2 in predicting the left ventricle remodeling six months after ST-segment elevation myocardial infarction. PATIENTS AND METHODS Materials and methods: The study involved 134 ST-segment elevation myocardial infarction patients. Occurrence of post-percutaneous coronary (PCI) intervention epicardial blood flow of TIMI <3 or myocardial blush grade 0-1 along with ST resolution <70% within 2 hours after PCI was qualified as the no-reflow condition. Left ventricle remodeling was defined after 6-months as an increase in left ventricle end-diastolic volume and/or end-systolic volume by more than 10%. RESULTS Results: A logistic regression formula was evaluated. Included biomarkers were macrophage migration inhibitory factor and sST2, left ventricle ejection fraction: Y=exp(-39.06+0.82EF+0.096ST2+0.0028MIF) / (1+exp(-39.06+0.82EF+0.096ST2+0.0028MIF)). The estimated range is from 0 to 1 point. Less than 0.5 determines an adverse outcome, and more than 0.5 is a good prognosis. This equation, with sensitivity of 77 % and specificity of 85%, could predict the development of adverse left ventricle remodeling six months after a coronary event (AUC=0.864, CI 0.673 to 0.966, p<0.05). CONCLUSION Conclusions: A combination of biomarkers gives a significant predicting result in the formation of adverse left ventricular remodeling after ST-segment elevation myocardial infarction.
Collapse
Affiliation(s)
- Iryna R Vyshnevska
- GOVERNMENT INSTITUTION "LT MALAYA THERAPY NATIONAL INSTITUTE OF THE NAMS OF UKRAINE", KHARKIV, UKRAINE
| | - Tatyana Storozhenko
- GOVERNMENT INSTITUTION "LT MALAYA THERAPY NATIONAL INSTITUTE OF THE NAMS OF UKRAINE", KHARKIV, UKRAINE
| | - Mykola P Kopytsya
- GOVERNMENT INSTITUTION "LT MALAYA THERAPY NATIONAL INSTITUTE OF THE NAMS OF UKRAINE", KHARKIV, UKRAINE
| | - Natalia V Bila
- V.N. KARAZIN KHARKIV NATIONAL UNIVERSITY, KHARKIV, UKRAINE
| | - Andrii Kis
- V.N. KARAZIN KHARKIV NATIONAL UNIVERSITY, KHARKIV, UKRAINE
| | - Mohammad Kaaki
- V.N. KARAZIN KHARKIV NATIONAL UNIVERSITY, KHARKIV, UKRAINE
| |
Collapse
|
4
|
Changes in the Expression of MIF and Other Key Enzymes of Energy Metabolism in the Myocardia of Broiler Chickens with Ascites Syndrome. Animals (Basel) 2022; 12:ani12192488. [PMID: 36230229 PMCID: PMC9558964 DOI: 10.3390/ani12192488] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/09/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022] Open
Abstract
Ascites syndrome (AS) is a metabolic disease observed mainly in fast-growing broilers. The heart is one of the most important target organs of the disease. The goal of this study was to evaluate the metabolic function of the right ventricles in clinical ascitic broilers. HE staining was performed to observe histopathological changes in the right ventricle of the heart, while Western blotting was used to detect the protein expression levels of macrophage migration inhibitory factor (MIF) and phosphorylated AMP-activated protein kinase (p-AMPK), as well as other key enzymes of energy metabolic pathways (i.e., glycolytic pathway: HK2, PFK1, PFK2, and PKM2; the tricarboxylic acid cycle (TCA cycle) pathway: OGDH, IDH2, and CS; and the fatty acid oxidation pathway: CPT-1A and ACC) in myocardial tissue. The histopathological examination of the myocardia of ascitic broilers revealed disoriented myocardial cells in the myofibril structure and a large number of blood cells deposited in the intermyofibrillar vessels, suggesting right heart failure in ascitic broilers. The Western blotting analysis demonstrated significantly increased levels of MIF and p-AMPK in the myocardia of ascitic broilers compared to those of the control group (p < 0.05). Additionally, the protein expression of key enzymes was dramatically increased in the glycolytic and fatty acid oxidation pathways, while the protein expression of key enzymes in the TCA cycle pathway was decreased in the ascitic broiler group. These findings suggest enhanced glycolysis and fatty acid oxidation metabolism, and a diminished TCA cycle, in the myocardia of broiler chickens with ascites syndrome.
Collapse
|
5
|
Obstructive Sleep Apnea Syndrome In Vitro Model: Controlled Intermittent Hypoxia Stimulation of Human Stem Cells-Derived Cardiomyocytes. Int J Mol Sci 2022; 23:ijms231810272. [PMID: 36142186 PMCID: PMC9499466 DOI: 10.3390/ijms231810272] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
Cardiovascular morbidity is the leading cause of death of obstructive sleep apnea (OSA) syndrome patients. Nocturnal airway obstruction is associated with intermittent hypoxia (IH). In our previous work with cell lines, incubation with sera from OSA patients induced changes in cell morphology, NF-κB activation and decreased viability. A decrease in beating rate, contraction amplitude and a reduction in intracellular calcium signaling was also observed in human cardiomyocytes differentiated from human embryonic stem cells (hESC-CMs). We expanded these observations using a new controlled IH in vitro system on beating hESC-CMs. The Oxy-Cycler system was programed to generate IH cycles. Following IH, we detected the activation of Hif-1α as an indicator of hypoxia and nuclear NF-κB p65 and p50 subunits, representing pro-inflammatory activity. We also detected the secretion of inflammatory cytokines, such as MIF, PAI-1, MCP-1 and CXCL1, and demonstrated a decrease in beating rate of hESC-CMs following IH. IH induces the co-activation of inflammatory features together with cardiomyocyte alterations which are consistent with myocardial damage in OSA. This study provides an innovative approach for in vitro studies of OSA cardiovascular morbidity and supports the search for new pharmacological agents and molecular targets to improve diagnosis and treatment of patients.
Collapse
|
6
|
Skeens E, Gadzuk-Shea M, Shah D, Bhandari V, Schweppe DK, Berlow RB, Lisi GP. Redox-dependent structure and dynamics of macrophage migration inhibitory factor reveal sites of latent allostery. Structure 2022; 30:840-850.e6. [PMID: 35381187 DOI: 10.1016/j.str.2022.03.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/20/2022] [Accepted: 03/09/2022] [Indexed: 01/01/2023]
Abstract
Macrophage migration inhibitory factor (MIF) is a multifunctional immunoregulatory protein that is a key player in the innate immune response. Given its overexpression at sites of inflammation and in diseases marked by increasingly oxidative environments, a comprehensive understanding of how cellular redox conditions impact the structure and function of MIF is necessary. We used NMR spectroscopy and mass spectrometry to investigate biophysical signatures of MIF under varied solution redox conditions. Our results indicate that the MIF structure is modified and becomes increasingly dynamic in an oxidative environment, which may be a means to alter the MIF conformation and functional response in a redox-dependent manner. We identified latent allosteric sites within MIF through mutational analysis of redox-sensitive residues, revealing that a loss of redox-responsive residues attenuates CD74 receptor activation. Leveraging sites of redox sensitivity as targets for structure-based drug design therefore reveals an avenue to modulate MIF function in its "disease state."
Collapse
Affiliation(s)
- Erin Skeens
- Department of Molecular Biology, Cell Biology, & Biochemistry, Brown University, Providence, RI 02903, USA
| | - Meagan Gadzuk-Shea
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Dilip Shah
- Section of Neonatology, Department of Pediatrics, Cooper University Hospital, Camden, NJ 08103, USA
| | - Vineet Bhandari
- Section of Neonatology, Department of Pediatrics, Cooper University Hospital, Camden, NJ 08103, USA
| | - Devin K Schweppe
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Rebecca B Berlow
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Biochemistry and Biophysics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - George P Lisi
- Department of Molecular Biology, Cell Biology, & Biochemistry, Brown University, Providence, RI 02903, USA.
| |
Collapse
|
7
|
Mravec Bencúrová D, Mandíková Š, Daňková P. Age and sex-specific differences in interleukin 4, interferon gamma, macrophage migration inhibitory factor, and vascular endothelial growth factor levels in the tears of healthy subjects. Eur J Ophthalmol 2021; 32:1900-1906. [PMID: 34825591 DOI: 10.1177/11206721211064023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To investigate the physiological profile of pro-inflammatory and anti-inflammatory cytokines in tears produced by epithelial cells under the effect of endogenous and exogenous biological factors. Knowing the physiological cytokine profile in tears with its biological characteristics including sex- and age-specific effects is fundamental when tears are analyzed for diagnostic or prognostic purposes in eye diseases. METHODS Tear samples were collected from right eye of 45 healthy volunteers (24 males, 21 females) by 5 μl microcapillary tube. Cytokines interleukin 1β, interleukin 10, interleukin 4, interferon gamma, macrophage migration inhibitory factor, and vascular endothelial growth factor were quantified by multiplex Bio-Plex system. RESULTS The production of macrophage migration inhibitory factor cytokine by epithelial cells on the ocular surface is higher in males compared to females (p = 0.05); actually, most of female tear samples present with undetectable macrophage migration inhibitory factor levels. Our results show the negative correlations between the age and concentrations of interleukin 4 (p < 0.01) and interferon gamma (p < 0.01) in tears, respectively, and positive associations of vascular endothelial growth factor levels with the age above 45 years (p < 0.05). CONCLUSIONS Data in this study indicate that age and sex may affect the physiological levels of cytokines in tears. Consequently, the impacts of biological factors need to be recognized and taken into consideration before the levels of cytokines in patients' tears are analyzed for medical reasons. Concentrations of interleukin 1β and interleukin 10 cytokines, however, are very low in healthy tears and do not seem to be influenced by studied biological factors; therefore, they meet the requirements for analytes suitable for medical diagnostic and prognostic purposes.
Collapse
Affiliation(s)
- Dominika Mravec Bencúrová
- Department of Anthropology and Human Genetics, Faculty of Science, 112302Charles University, Prague, Czech Republic
| | - Šárka Mandíková
- Department of Anthropology and Human Genetics, Faculty of Science, 112302Charles University, Prague, Czech Republic
| | - Pavlína Daňková
- Department of Anthropology and Human Genetics, Faculty of Science, 112302Charles University, Prague, Czech Republic
| |
Collapse
|
8
|
Daiber A, Steven S, Euler G, Schulz R. Vascular and Cardiac Oxidative Stress and Inflammation as Targets for Cardioprotection. Curr Pharm Des 2021; 27:2112-2130. [PMID: 33550963 DOI: 10.2174/1381612827666210125155821] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 11/11/2020] [Indexed: 11/22/2022]
Abstract
Cardiac and vascular diseases are often associated with increased oxidative stress and inflammation, and both may contribute to the disease progression. However, successful applications of antioxidants in the clinical setting are very rare and specific anti-inflammatory therapeutics only emerged recently. Reasons for this rely on the great diversity of oxidative stress and inflammatory cells that can either act as cardioprotective or cause tissue damage in the heart. Recent large-scale clinical trials found that highly specific anti-inflammatory therapies using monoclonal antibodies against cytokines resulted in lower cardiovascular mortality in patients with pre-existing atherosclerotic disease. In addition, unspecific antiinflammatory medication and established cardiovascular drugs with pleiotropic immunomodulatory properties such as angiotensin converting enzyme (ACE) inhibitors or statins have proven beneficial cardiovascular effects. Normalization of oxidative stress seems to be a common feature of these therapies, which can be explained by a close interaction/crosstalk of the cellular redox state and inflammatory processes. In this review, we give an overview of cardiac reactive oxygen species (ROS) sources and processes of cardiac inflammation as well as the connection of ROS and inflammation in ischemic cardiomyopathy in order to shed light on possible cardioprotective interventions.
Collapse
Affiliation(s)
- Andreas Daiber
- Department of Cardiology, Molecular Cardiology, University Medical Center Mainz, Mainz, Germany
| | - Sebastian Steven
- Department of Cardiology, Molecular Cardiology, University Medical Center Mainz, Mainz, Germany
| | - Gerhild Euler
- Institute of Physiology, Justus-Liebig University, Giessen, Germany
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University, Giessen, Germany
| |
Collapse
|
9
|
Luo JY, Fang BB, Du GL, Liu F, Li YH, Tian T, Li XM, Gao XM, Yang YN. Association between MIF gene promoter rs755622 and susceptibility to coronary artery disease and inflammatory cytokines in the Chinese Han population. Sci Rep 2021; 11:8050. [PMID: 33850223 PMCID: PMC8044220 DOI: 10.1038/s41598-021-87580-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 03/17/2021] [Indexed: 02/01/2023] Open
Abstract
Macrophage migration inhibitory factor (MIF) is an essential mediator of atherosclerotic plaque progression and instability leading to intracoronary thrombosis, therefore contributing to coronary artery disease (CAD). In this study, we investigated the relationship between MIF gene polymorphism and CAD in Chinese Han population. Three single nucleotide polymorphisms (SNP, rs755622, rs1007888 and rs2096525) of MIF gene were genotyped by TaqMan genotyping assay in 1120 control participants and 1176 CAD patients. Coronary angiography was performed in all CAD patients and Gensini score was used to assess the severity of coronary artery lesions. The plasma levels of MIF and other inflammatory mediators were measured by ELISA. The CAD patients had a higher frequency of CC genotype and C allele of rs755622 compared with that in control subjects (CC genotype: 6.5% vs. 3.9%, P = 0.008, C allele: 24.0% vs. 20.6%, P = 0.005). The rs755622 CC genotype was associated with an increased risk of CAD (OR: 1.804, 95%CI: 1.221-2.664, P = 0.003). CAD patients with a variation of rs755622 CC genotype had significantly higher Gensini score compared with patients with GG or CG genotype (all P < 0.05). In addition, the circulating MIF level was highest in CAD patients carrying rs755622 CC genotype (40.7 ± 4.2 ng/mL) and then followed by GC (37.9 ± 3.4 ng/mL) or GG genotype (36.9 ± 3.7 ng/mL, all P < 0.01). Our study showed an essential relationship between the MIF gene rs755622 variation and CAD in Chinese Han population. Individuals who carrying MIF gene rs755622 CC genotype were more susceptible to CAD and had more severe coronary artery lesion. This variation also had a potential influence in circulating MIF levels.
Collapse
Affiliation(s)
- Jun-Yi Luo
- 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, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China
| | - Bin-Bin Fang
- 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, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China
| | - Guo-Li Du
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 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, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China
| | - Yan-Hong Li
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China
- Department of Clinical Laboratory, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Ting Tian
- 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, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Clinical Medical Research Institute 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, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, 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, 137 Liyushan South Road, Urumqi, 830054, China.
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China.
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, 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, 137 Liyushan South Road, Urumqi, 830054, China.
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Clinical Medical Research Institute of Xinjiang Medical University, Urumqi, China.
| |
Collapse
|
10
|
Illescas O, Pacheco-Fernández T, Laclette JP, Rodriguez T, Rodriguez-Sosa M. Immune modulation by the macrophage migration inhibitory factor (MIF) family: D-dopachrome tautomerase (DDT) is not (always) a backup system. Cytokine 2020; 133:155121. [PMID: 32417648 DOI: 10.1016/j.cyto.2020.155121] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 04/29/2020] [Accepted: 05/06/2020] [Indexed: 01/06/2023]
Abstract
Human macrophage migration inhibition factor (MIF) is a protein with cytokine and chemokine properties that regulates a diverse range of physiological functions related to innate immunity and inflammation. Most research has focused on the role of MIF in different inflammatory diseases. D-dopachrome tautomerase (DDT), a different molecule with structural similarities to MIF, which shares receptors and biological functions, has recently been reported, but little is known about its roles and mechanisms. In this review, we sought to understand the similarities and differences between these molecules by summarizing what is known about their different structures, receptors and mechanisms regulating their expression and biological activities with an emphasis on immunological aspects.
Collapse
Affiliation(s)
- Oscar Illescas
- Biomedicine Unit, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México (UNAM), Tlalnepantla, MEX C.P. 54090, Mexico
| | - Thalia Pacheco-Fernández
- Biomedicine Unit, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México (UNAM), Tlalnepantla, MEX C.P. 54090, Mexico
| | - Juan P Laclette
- Department of Immunology, Institute of Biomedical Research, Universidad Nacional Autónoma de México (UNAM), Mexico City C.P. 04510, Mexico
| | - Tonathiu Rodriguez
- Biomedicine Unit, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México (UNAM), Tlalnepantla, MEX C.P. 54090, Mexico
| | - Miriam Rodriguez-Sosa
- Biomedicine Unit, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México (UNAM), Tlalnepantla, MEX C.P. 54090, Mexico.
| |
Collapse
|
11
|
Jankauskas SS, Wong DW, Bucala R, Djudjaj S, Boor P. Evolving complexity of MIF signaling. Cell Signal 2019; 57:76-88. [DOI: 10.1016/j.cellsig.2019.01.006] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 01/17/2019] [Accepted: 01/18/2019] [Indexed: 01/27/2023]
|
12
|
Voss S, Krüger S, Scherschel K, Warnke S, Schwarzl M, Schrage B, Girdauskas E, Meyer C, Blankenberg S, Westermann D, Lindner D. Macrophage Migration Inhibitory Factor (MIF) Expression Increases during Myocardial Infarction and Supports Pro-Inflammatory Signaling in Cardiac Fibroblasts. Biomolecules 2019; 9:biom9020038. [PMID: 30678084 PMCID: PMC6406883 DOI: 10.3390/biom9020038] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/11/2019] [Accepted: 01/17/2019] [Indexed: 01/01/2023] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine known to play a major role in inflammatory diseases such as myocardial infarction (MI), where its expression increases. Cardio-protective functions of MIF during ischemia have been reported. Recently, the structurally related MIF-2 was identified and similar effects are assumed. We wanted to further investigate the role of MIF and MIF-2 on inflammatory processes during MI. Therefore, we subjected mice to experimentally induced MI by coronary occlusion for one and five days. During the acute phase of MI, the gene expression of Mif was upregulated in the infarct zone, whereas Mif-2 was downregulated, suggesting a minor role of MIF-2. Simulating ischemic conditions or mechanical stress in vitro, we demonstrated that Mif expression was induced in resident cardiac cells. To investigate possible auto-/paracrine effects, cardiomyocytes and cardiac fibroblasts were individually treated with recombinant murine MIF, which in turn induced Mif expression and the expression of pro-inflammatory genes in cardiac fibroblasts. Cardiomyocytes did not respond to recombinant MIF with pro-inflammatory gene expression. While MIF stimulation alone did not change the expression of pro-fibrotic genes in cardiac fibroblasts, ischemia reduced their expression. Mimicking the increased MIF levels during MI, we exposed cardiac fibroblasts to simulated ischemia in the presence of MIF, which led to further reduced expression of pro-fibrotic genes. The presented data show that MIF was expressed by resident cardiac cells during MI. In vitro, Mif expression was induced by different external stimuli in cardiomyocytes and cardiac fibroblasts. Addition of recombinant MIF protein increased the expression of pro-inflammatory genes in cardiac fibroblasts including Mif expression itself. Thereby, cardiac fibroblasts may amplify Mif expression during ischemia promoting cardiomyocyte survival.
Collapse
Affiliation(s)
- Svenja Voss
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany.
- Partner Site Hamburg/Kiel/Lübeck, DZHK (German Center for Cardiovascular Research), 20246 Hamburg, Germany.
| | - Saskia Krüger
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany.
- Partner Site Hamburg/Kiel/Lübeck, DZHK (German Center for Cardiovascular Research), 20246 Hamburg, Germany.
| | - Katharina Scherschel
- Partner Site Hamburg/Kiel/Lübeck, DZHK (German Center for Cardiovascular Research), 20246 Hamburg, Germany.
- Clinic for Cardiology-Electrophysiology, University Heart Center Hamburg, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Svenja Warnke
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Michael Schwarzl
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany.
- Partner Site Hamburg/Kiel/Lübeck, DZHK (German Center for Cardiovascular Research), 20246 Hamburg, Germany.
| | - Benedikt Schrage
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany.
- Partner Site Hamburg/Kiel/Lübeck, DZHK (German Center for Cardiovascular Research), 20246 Hamburg, Germany.
| | - Evaldas Girdauskas
- Partner Site Hamburg/Kiel/Lübeck, DZHK (German Center for Cardiovascular Research), 20246 Hamburg, Germany.
- Clinic for Cardiovascular Surgery, University Heart Center Hamburg, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Christian Meyer
- Partner Site Hamburg/Kiel/Lübeck, DZHK (German Center for Cardiovascular Research), 20246 Hamburg, Germany.
- Clinic for Cardiology-Electrophysiology, University Heart Center Hamburg, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Stefan Blankenberg
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany.
- Partner Site Hamburg/Kiel/Lübeck, DZHK (German Center for Cardiovascular Research), 20246 Hamburg, Germany.
| | - Dirk Westermann
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany.
- Partner Site Hamburg/Kiel/Lübeck, DZHK (German Center for Cardiovascular Research), 20246 Hamburg, Germany.
| | - Diana Lindner
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany.
- Partner Site Hamburg/Kiel/Lübeck, DZHK (German Center for Cardiovascular Research), 20246 Hamburg, Germany.
| |
Collapse
|
13
|
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: 0.7] [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
|
14
|
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: 58] [Impact Index Per Article: 7.3] [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
|
15
|
Ochi A, Chen D, Schulte W, Leng L, Moeckel N, Piecychna M, Averdunk L, Stoppe C, Bucala R, Moeckel G. MIF-2/D-DT enhances proximal tubular cell regeneration through SLPI- and ATF4-dependent mechanisms. Am J Physiol Renal Physiol 2017; 313:F767-F780. [PMID: 28539339 DOI: 10.1152/ajprenal.00683.2016] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 05/23/2017] [Accepted: 05/24/2017] [Indexed: 12/29/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a cytokine with pleiotropic actions that is produced by several organs and cell types. Depending on the target cell and the inflammatory context, MIF can engage its two component receptor complex CD74 and CD44 and the chemokine receptors CXCR2/4. MIF is constitutively expressed in renal proximal tubular cells, stored in intracellular preformed pools, and released at a low rate. Recently, a second MIF-like protein (i.e., MIF-2/D-DT) has been characterized in mammals. Our study was aimed at examining the role of MIF-2/D-DT, which mediates tissue protection in the heart, in tubular cell regeneration from ischemia-reperfusion injury. We found that Mif-/-, Mif-2-/-, and Cd74-/- mice had significantly worse tubular injury compared with wild-type (WT) control mice and that treatment with MIF-2/D-DT significantly improved recovery of injured epithelial cells. RNAseq analysis of kidney tissue from the ischemia-reperfusion injury model revealed that MIF-2/D-DT treatment stimulates secretory leukocyte proteinase inhibitor (SLPI) and cyclin D1 expression. MIF-2/D-DT additionally activates of eukaryotic initiation factor (eIF) 2α and activating transcription factor (ATF) 4, two transcription factors involved in the integrated stress response (ISR), which is a cellular stress response activated by hypoxia, nutrient deprivation, and oxygen radicals. MIF-2/D-DT also inhibited apoptosis and induced autophagy in hypoxia-treated mouse proximal tubular (MPT) cells. These results indicate that MIF-2/D-DT is an important factor in tubular cell regeneration and may be of therapeutic utility as a regenerative agent in the clinical setting of ischemic acute kidney injury.
Collapse
Affiliation(s)
- Akinobu Ochi
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Dong Chen
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Wibke Schulte
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut; and
| | - Lin Leng
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut; and
| | - Nickolas Moeckel
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Marta Piecychna
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut; and
| | - Luisa Averdunk
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut.,Department of Anesthesiology and Intensive Care Medicine, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Christian Stoppe
- Department of Anesthesiology and Intensive Care Medicine, Rheinisch-Westfälische Technische Hochschule Aachen, Aachen, Germany
| | - Richard Bucala
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut; and
| | - Gilbert Moeckel
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut;
| |
Collapse
|
16
|
Pohl J, Hendgen-Cotta UB, Stock P, Luedike P, Rassaf T. Elevated MIF-2 levels predict mortality in critically ill patients. J Crit Care 2017; 40:52-57. [PMID: 28329734 DOI: 10.1016/j.jcrc.2017.03.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 02/05/2017] [Accepted: 03/12/2017] [Indexed: 12/11/2022]
Abstract
PURPOSE D-dopachrome tautomerase (MIF-2 or DDT) is a member of the macrophage migration inhibitory factor (MIF) superfamily and a close structural homolog to MIF. Circulating MIF-2 has been described to be elevated in patients suffering from sepsis, severe burn injury and after surgery. We sought to evaluate the prognostic value of MIF-2 in critically ill patients. METHODS A total of 72 patients were studied upon admission to the medical intensive care unit (ICU). MIF and MIF-2 levels were assessed and compared to healthy controls. Clinical data, various laboratory parameters and mortality were assessed. RESULTS We found significantly elevated levels of MIF-2 and MIF at admission to the ICU in critically ill patients compared to healthy controls. MIF-2 levels were associated with disease severity as measured by APACHE II scores. MIF-2 levels in ICU patients correlated with biomarkers reflecting organ damage, but were not influenced by acute or chronic kidney disease. Kaplan-Meier analysis revealed distinctly elevated mortality in patients with high plasma MIF-2 levels. CONCLUSIONS MIF-2 levels are elevated in critically ill patients and linked to parameters of organ damage, supporting its value as a potential tool for the assessment of prognosis in critical illness.
Collapse
Affiliation(s)
- Julia Pohl
- University Hospital Essen, West German Heart and Vascular Center, Department of Cardiology and Vascular Medicine, Hufelandstr. 55, 45147 Essen, Germany.
| | - Ulrike B Hendgen-Cotta
- University Hospital Essen, West German Heart and Vascular Center, Department of Cardiology and Vascular Medicine, Hufelandstr. 55, 45147 Essen, Germany
| | - Pia Stock
- University Hospital Essen, West German Heart and Vascular Center, Department of Cardiology and Vascular Medicine, Hufelandstr. 55, 45147 Essen, Germany
| | - Peter Luedike
- University Hospital Essen, West German Heart and Vascular Center, Department of Cardiology and Vascular Medicine, Hufelandstr. 55, 45147 Essen, Germany
| | - Tienush Rassaf
- University Hospital Essen, West German Heart and Vascular Center, Department of Cardiology and Vascular Medicine, Hufelandstr. 55, 45147 Essen, Germany
| |
Collapse
|
17
|
Jiang W, Wang X, Osborne OJ, Du Y, Chang CH, Liao YP, Sun B, Jiang J, Ji Z, Li R, liu X, Lu J, Lin S, Meng H, Xia T, Nel AE. Pro-Inflammatory and Pro-Fibrogenic Effects of Ionic and Particulate Arsenide and Indium-Containing Semiconductor Materials in the Murine Lung. ACS NANO 2017; 11:1869-1883. [PMID: 28177603 PMCID: PMC5543990 DOI: 10.1021/acsnano.6b07895] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
We have recently shown that the toxicological potential of GaAs and InAs particulates in cells is size- and dissolution-dependent, tending to be more pronounced for nano- vs micron-sized particles. Whether the size-dependent dissolution and shedding of ionic III-V materials also apply to pulmonary exposure is unclear. While it has been demonstrated that micron-sized III-V particles, such as GaAs and InAs, are capable of inducing hazardous pulmonary effects in an occupational setting as well as in animal studies, the effect of submicron particles (e.g., the removal of asperities during processing of semiconductor wafers) is unclear. We used cytokine profiling to compare the pro-inflammatory effects of micron- and nanoscale GaAs and InAs particulates in cells as well as the murine lung 40 h and 21 days after oropharyngeal aspiration. Use of cytokine array technology in macrophage and epithelial cell cultures demonstrated a proportionally higher increase in the levels of matrix metalloproteinase inducer (EMMPRIN), macrophage migration inhibitory factor (MIF), and interleukin 1β (IL-1β) by nanosized (n) GaAs and n-InAs as well as As(III). n-GaAs and n-InAs also triggered higher neutrophil counts in the bronchoalveolar lavage fluid (BALF) of mice than micronscale particles 40 h post-aspiration, along with increased production of EMMPRIN and MIF. In contrast, in animals sacrificed 21 days after exposure, only n-InAs induced fibrotic lung changes as determined by increased lung collagen as well as increased levels of TGF-β1 and PDGF-AA in the BALF. A similar trend was seen for EMMPRIN and matrix metallopeptidase (MMP-9) levels in the BALF. Nano- and micron-GaAs had negligible subacute effects. Importantly, the difference between the 40 h and 21 days data appears to be biopersistence of n-InAs, as demonstrated by ICP-OES analysis of lung tissue. Interestingly, an ionic form of In, InCl3, also showed pro-fibrogenic effects due to the formation of insoluble In(OH)3 nanostructures. All considered, these data indicate that while nanoscale particles exhibit increased pro-inflammatory effects in the lung, most effects are transient, except for n-InAs and insoluble InCl3 species that are biopersistent and trigger pro-fibrotic effects. These results are of potential importance for the understanding the occupational health effects of III-V particulates.
Collapse
Affiliation(s)
- Wen Jiang
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, 570 Westwood Plaza, Los Angeles, CA 90095, United States
| | - Xiang Wang
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, 570 Westwood Plaza, Los Angeles, CA 90095, United States
| | - Olivia J. Osborne
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, 570 Westwood Plaza, Los Angeles, CA 90095, United States
| | - Yingjie Du
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, 570 Westwood Plaza, Los Angeles, CA 90095, United States
| | - Chong Hyun Chang
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, 570 Westwood Plaza, Los Angeles, CA 90095, United States
| | - Yu-Pei Liao
- Division of NanoMedicine, Department of Medicine, University of California Los Angeles, 10833 Le Conte Ave, Los Angeles, CA 90095, United States
| | - Bingbing Sun
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, 570 Westwood Plaza, Los Angeles, CA 90095, United States
| | - Jinhong Jiang
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, 570 Westwood Plaza, Los Angeles, CA 90095, United States
| | - Zhaoxia Ji
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, 570 Westwood Plaza, Los Angeles, CA 90095, United States
| | - Ruibin Li
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, 570 Westwood Plaza, Los Angeles, CA 90095, United States
- School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Medical College of Soochow University, Suzhou, Jiangsu 215123, China
| | - Xiangsheng liu
- Division of NanoMedicine, Department of Medicine, University of California Los Angeles, 10833 Le Conte Ave, Los Angeles, CA 90095, United States
| | - Jianqin Lu
- Division of NanoMedicine, Department of Medicine, University of California Los Angeles, 10833 Le Conte Ave, Los Angeles, CA 90095, United States
| | - Sijie Lin
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, 570 Westwood Plaza, Los Angeles, CA 90095, United States
- College of Environmental Science and Engineering State Key Laboratory of Pollution Control and Resource Reuse, Tongji University, Shanghai, China, 200092
| | - Huan Meng
- Division of NanoMedicine, Department of Medicine, University of California Los Angeles, 10833 Le Conte Ave, Los Angeles, CA 90095, United States
| | - Tian Xia
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, 570 Westwood Plaza, Los Angeles, CA 90095, United States
- Division of NanoMedicine, Department of Medicine, University of California Los Angeles, 10833 Le Conte Ave, Los Angeles, CA 90095, United States
| | - André E. Nel
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute, University of California Los Angeles, 570 Westwood Plaza, Los Angeles, CA 90095, United States
- Division of NanoMedicine, Department of Medicine, University of California Los Angeles, 10833 Le Conte Ave, Los Angeles, CA 90095, United States
- Address correspondence to: André E. Nel, M.D./Ph.D., Department of Medicine, Division of NanoMedicine, UCLA School of Medicine, 52-175 CHS, 10833 Le Conte Ave, Los Angeles, CA 90095-1680, USA, Tel: (310) 825-6620, Fax: (310) 206-8107,
| |
Collapse
|
18
|
Al-Mohanna F. The Cardiokines. ENDOCRINOLOGY OF THE HEART IN HEALTH AND DISEASE 2017:87-114. [DOI: 10.1016/b978-0-12-803111-7.00004-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
19
|
Fan F, Fang L, Moore XL, Xie X, Du XJ, White DA, O'Brien J, Thomson H, Wang J, Schneider HG, Ellims A, Barber TW, Dart AM. Plasma Macrophage Migration Inhibitor Factor Is Elevated in Response to Myocardial Ischemia. J Am Heart Assoc 2016; 5:JAHA.115.003128. [PMID: 27364992 PMCID: PMC5015363 DOI: 10.1161/jaha.115.003128] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Macrophage migration inhibitory factor (MIF) is a key regulator of inflammatory responses, including in the heart. Plasma MIF is elevated early in the course of acute myocardial infarction. In this study, we hypothesized that plasma MIF may also be increased in acute myocardial ischemia. METHODS AND RESULTS Patients undergoing cardiac stress test (stress nuclear myocardial perfusion scan or stress echocardiography) were recruited. Twenty-two patients had a stress test indicative of myocardial ischemia and were compared with 62 patients who had a negative stress test. Plasma MIF was measured by ELISA before and after the stress test. MIF was also measured in patients with peripheral arterial occlusive disease before and after exercise causing claudication. Gene and protein expression of MIF was measured in mouse cardiac and skeletal muscle tissue by real-time polymerase chain reaction and western blot, respectively. Plasma MIF was elevated at 5 and 15 minutes after stress (relative to before stress) in patients with a positive test, compared with those with a negative test. In contrast, high-sensitivity troponin T and C-reactive protein were not altered after stress in either group. MIF was not altered after exercise in PAOD patients, despite the occurrence of claudication, suggesting that plasma MIF is not a marker for skeletal muscle ischemia. This may be explained by a lower gene and protein expression of MIF in skeletal muscle than the heart. CONCLUSIONS Our results suggest that plasma MIF is an early marker for acute myocardial ischemia.
Collapse
Affiliation(s)
- Fenling Fan
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia Department of Cardiovascular Medicine, The 1st Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, China
| | - Lu Fang
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Xiao-Lei Moore
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Xuegang Xie
- Department of Cardiovascular Medicine, The 1st Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, China
| | - Xiao-Jun Du
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - David A White
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Jessica O'Brien
- Department of Cardiovascular Medicine, The Alfred Hospital, Melbourne, Victoria, Australia
| | - Helen Thomson
- Department of Cardiovascular Medicine, The Alfred Hospital, Melbourne, Victoria, Australia
| | - Jun Wang
- Department of Cardiovascular Medicine, The 1st Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, China
| | - Hans G Schneider
- Department of Chemical Pathology, The Alfred Hospital, Melbourne, Victoria, Australia
| | - Andris Ellims
- Department of Cardiovascular Medicine, The Alfred Hospital, Melbourne, Victoria, Australia
| | - Thomas W Barber
- Department of Nuclear Medicine, The Alfred Hospital, Melbourne, Victoria, Australia
| | - Anthony M Dart
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia Department of Cardiovascular Medicine, The Alfred Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
20
|
Wu J, Stefaniak J, Hafner C, Schramel JP, Kaun C, Wojta J, Ullrich R, Tretter VE, Markstaller K, Klein KU. Intermittent Hypoxia Causes Inflammation and Injury to Human Adult Cardiac Myocytes. Anesth Analg 2016; 122:373-80. [PMID: 26505576 DOI: 10.1213/ane.0000000000001048] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Intermittent hypoxia may occur in a number of clinical scenarios, including interruption of myocardial blood flow or breathing disorders such as obstructive sleep apnea. Although intermittent hypoxia has been linked to cardiovascular and cerebrovascular disease, the effect of intermittent hypoxia on the human heart is not fully understood. Therefore, in the present study, we compared the cellular responses of cultured human adult cardiac myocytes (HACMs) exposed to intermittent hypoxia and different conditions of continuous hypoxia and normoxia. METHODS HACMs were exposed to intermittent hypoxia (0%-21% O2), constant mild hypoxia (10% O2), constant severe hypoxia (0% O2), or constant normoxia (21% O2), using a novel cell culture bioreactor with gas-permeable membranes. Cell proliferation, lactate dehydrogenase release, vascular endothelial growth factor release, and cytokine (interleukin [IL] and macrophage migration inhibitory factor) release were assessed at baseline and after 8, 24, and 72 hours of exposure. A signal transduction pathway finder array was performed to determine the changes in gene expression. RESULTS In comparison with constant normoxia and constant mild hypoxia, intermittent hypoxia induced earlier and greater inflammatory response and extent of cell injury as evidenced by lower cell numbers and higher lactate dehydrogenase, vascular endothelial growth factor, and proinflammatory cytokine (IL-1β, IL-6, IL-8, and macrophage migration inhibitory factor) release. Constant severe hypoxia showed more detrimental effects on HACMs at later time points. Pathway analysis demonstrated that intermittent hypoxia primarily altered gene expression in oxidative stress, Wnt, Notch, and hypoxia pathways. CONCLUSIONS Intermittent and constant severe hypoxia, but not constant mild hypoxia or normoxia, induced inflammation and cell injury in HACMs. Cell injury occurred earliest and was greatest after intermittent hypoxia exposure. Our in vitro findings suggest that intermittent hypoxia exposure may produce rapid and substantial damage to the human heart.
Collapse
Affiliation(s)
- Jing Wu
- From the *Department of Anesthesia, General Intensive Care and Pain Management, Medical University of Vienna, Vienna, Austria; †Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; ‡Unit of Anesthesiology and Perioperative Intensive Care, University of Veterinary Medicine, Vienna, Austria; §Department of Internal Medicine II and ‖Core Facilities, Medical University of Vienna, Vienna, Austria; and ¶Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Xia W, Hou M. Macrophage migration inhibitory factor induces autophagy to resist hypoxia/serum deprivation-induced apoptosis via the AMP-activated protein kinase/mammalian target of rapamycin signaling pathway. Mol Med Rep 2016; 13:2619-26. [PMID: 26847932 DOI: 10.3892/mmr.2016.4847] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 10/23/2015] [Indexed: 11/05/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is an anti‑apoptotic agent in various cell types and protects the heart from stress‑induced injury by modulating autophagy. Autophagy, a conserved pathway for bulk degradation of intracellular proteins and organelles, helps to preserve and recycle energy and nutrients for cells to survive during starvation. The present study hypothesized that MIF protects bone marrow‑derived mesenchymal stem cells (MSCs) from apoptosis by modulating autophagy via the AMP‑activated protein kinase/mammalian target of rapamycin (AMPK/mTOR) signaling pathway. MSCs were obtained from rat bone marrow and cultured. Apoptosis was induced by hypoxia/serum deprivation for 24 h and was assessed using flow cytometry. MIF protected MSCs from apoptosis by modulating autophagy via the AMPK/mTOR signaling pathway resulting in increased expression of autophagy‑associated proteins (including LC3BI/LC3BII, Beclin‑1 and autophagy protein 5), and by increased phosphorylation of AMPK and decreased phosphorylation of mTOR. The MIF anti‑apoptotic effects were blocked by autophagy inhibitor, 3‑methyladenine or AMPK inhibitor, Compound C. These results indicate that MIF exerts a permissive role in protecting MSCs from apoptosis by regulation of autophagy via the AMPK/mTOR signaling pathway.
Collapse
Affiliation(s)
- Wenzheng Xia
- Department of Neurosurgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Meng Hou
- Department of Radiation Oncology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
22
|
Pohl J, Rammos C, Totzeck M, Stock P, Kelm M, Rassaf T, Luedike P. MIF reflects tissue damage rather than inflammation in post-cardiac arrest syndrome in a real life cohort. Resuscitation 2016; 100:32-7. [PMID: 26784134 DOI: 10.1016/j.resuscitation.2015.12.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 11/13/2015] [Accepted: 12/25/2015] [Indexed: 10/22/2022]
Abstract
INTRODUCTION Following successful resuscitation from cardiac arrest (CA), neurological impairment and other types of organ dysfunction cause significant morbidity and mortality-a condition termed post-cardiac arrest syndrome. Whole-body ischemia/reperfusion with oxygen debt activates immunologic and coagulation pathways increasing the risk of multiple organ failure and infection. We here examined the role of the pro-inflammatory cytokine macrophage migration inhibitory factor (MIF) in post-cardiac arrest syndrome. METHODS MIF plasma levels of n=16 patients with return of spontaneous circulation (ROSC) after CA were assessed with a previously validated method and compared to markers of systemic inflammation and cellular damage. ICU patients without former CA and healthy volunteers served as controls. RESULTS MIF levels in patients after ROSC were higher compared to those in healthy volunteers and ICU patients without CA. Kaplan-Meyer analysis revealed a distinctly elevated mortality since day one that further increased towards an elevated 60-days-mortality in patients with high plasma MIF. ROC curve identified plasma MIF as a predictor for mortality in patients after CA. Correlation with inflammatory parameters revealed that high MIF levels did not mirror post CA inflammatory syndrome, but distinctive cellular damage after ROSC as there were strong correlations with markers of cellular damage like LDH and GOT/GPT. CONCLUSION High MIF levels were associated with elevated 60-days-mortality and high MIF predicted mortality after CA. We found a close relation between circulating MIF levels and cellular damage, but not with an inflammatory syndrome.
Collapse
Affiliation(s)
- Julia Pohl
- University Hospital Essen, West German Heart and Vascular Center, Department of Cardiology, Hufelandstr. 55, Essen 45147, Germany
| | - Christos Rammos
- University Hospital Essen, West German Heart and Vascular Center, Department of Cardiology, Hufelandstr. 55, Essen 45147, Germany
| | - Matthias Totzeck
- University Hospital Essen, West German Heart and Vascular Center, Department of Cardiology, Hufelandstr. 55, Essen 45147, Germany
| | - Pia Stock
- University Hospital Essen, West German Heart and Vascular Center, Department of Cardiology, Hufelandstr. 55, Essen 45147, Germany
| | - Malte Kelm
- University Hospital Duesseldorf, Medical Faculty Heinrich-Heine-University, Division of Cardiology, Pulmonology and Vascular Medicine, Moorenstrasse 5, Duesseldorf D-40225, Germany
| | - Tienush Rassaf
- University Hospital Essen, West German Heart and Vascular Center, Department of Cardiology, Hufelandstr. 55, Essen 45147, Germany
| | - Peter Luedike
- University Hospital Essen, West German Heart and Vascular Center, Department of Cardiology, Hufelandstr. 55, Essen 45147, Germany.
| |
Collapse
|
23
|
Chesney JA, Mitchell RA. 25 Years On: A Retrospective on Migration Inhibitory Factor in Tumor Angiogenesis. Mol Med 2015; 21 Suppl 1:S19-24. [PMID: 26605643 DOI: 10.2119/molmed.2015.00055] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 03/16/2015] [Indexed: 01/26/2023] Open
Abstract
Twenty-five years ago marked the publication of the first report describing a functional contribution by the cytokine, macrophage migration inhibitory factor (MIF), to tumor-associated angiogenesis and growth. Since first appearing, this report has been cited 304 times (as of this writing), underscoring not only the importance of this landmark study but also the importance of MIF in tumor neovascularization. Perhaps more importantly, this first link between MIF and stromal cell-dependent tumor angiogenesis presaged the subsequent identification of MIF in mediating protumorigenic contributions to several solid tumor stromal cell types, including monocytes, macrophages, T lymphocytes, NK cells, fibroblasts, endothelial progenitors and mesenchymal stem cells. This retrospective review will broadly evaluate both past and present literature stemming from this initial publication, with an emphasis on cellular sources, cellular effectors, signal transduction mechanisms and the clinical importance of MIF-dependent tumor vascularization.
Collapse
Affiliation(s)
- Jason A Chesney
- Molecular Targets Program, JG Brown Cancer Center, and the Department of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Robert A Mitchell
- Molecular Targets Program, JG Brown Cancer Center, and the Department of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| |
Collapse
|
24
|
Pohl J, Hendgen-Cotta UB, Rammos C, Luedike P, Mull E, Stoppe C, Jülicher K, Lue H, Merx MW, Kelm M, Bernhagen J, Rassaf T. Targeted intracellular accumulation of macrophage migration inhibitory factor in the reperfused heart mediates cardioprotection. Thromb Haemost 2015; 115:200-12. [PMID: 26310191 DOI: 10.1160/th15-05-0436] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 07/09/2015] [Indexed: 01/05/2023]
Abstract
S-nitrosation of macrophage migration inhibitory factor (MIF) has been shown to be cytoprotective in myocardial ischaemia/reperfusion (I/R) injury. Since the exact mechanism of action is unknown, we here characterise the cardioprotective effects of targeted intracellular accumulation of MIF in myocardial I/R injury. We used different in vivo, ex vivo and in vitro models of myocardial I/R and hypoxia/reoxygenation (H/R) injury to determine MIF levels by immunoblots and ELISA in different phases of reperfusion and reoxygenation, respectively. We discovered a rapid decrease of cardiac MIF that was specific to the early phase of reperfusion. Posttranslational modification of MIF via S-nitrosation--proofed by a modified version of the Biotin Switch Assay--prevented this rapid decrease, leading to a targeted intracellular accumulation of MIF in the early phase of reperfusion. Intracellular MIF accumulation preserved the intracellular ability of MIF to reduce oxidative stress as shown by hydrogen peroxide and aconitase activity measurements. Infarct size measurements by TTC staining showed an overall enhanced cardioprotective effect of this protein by reduction of reperfusion injury. In summary, we have unravelled a novel mechanism of MIF-mediated cardioprotection. Targeted intracellular accumulation of MIF by S-nitrosation may offer a novel therapeutic approach in the treatment of myocardial I/R-injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Tienush Rassaf
- Prof. Dr. Tienush Rassaf, West-German Heart and Vascular Center Essen, Department of Cardiology, University Hospital Essen, Hufelandstr. 55, D-45147 Essen, Germany, Tel.: +49 201 723 4801, Fax: +49 201 723 5401, E-mail:
| |
Collapse
|
25
|
Systemic inflammatory response following acute myocardial infarction. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2015; 12:305-12. [PMID: 26089856 PMCID: PMC4460175 DOI: 10.11909/j.issn.1671-5411.2015.03.020] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 03/02/2015] [Accepted: 04/11/2015] [Indexed: 12/31/2022]
Abstract
Acute cardiomyocyte necrosis in the infarcted heart generates damage-associated molecular patterns, activating complement and toll-like receptor/interleukin-1 signaling, and triggering an intense inflammatory response. Inflammasomes also recognize danger signals and mediate sterile inflammatory response following acute myocardial infarction (AMI). Inflammatory response serves to repair the heart, but excessive inflammation leads to adverse left ventricular remodeling and heart failure. In addition to local inflammation, profound systemic inflammation response has been documented in patients with AMI, which includes elevation of circulating inflammatory cytokines, chemokines and cell adhesion molecules, and activation of peripheral leukocytes and platelets. The excessive inflammatory response could be caused by a deregulated immune system. AMI is also associated with bone marrow activation and spleen monocytopoiesis, which sustains a continuous supply of monocytes at the site of inflammation. Accumulating evidence has shown that systemic inflammation aggravates atherosclerosis and markers for systemic inflammation are predictors of adverse clinical outcomes (such as death, recurrent myocardial infarction, and heart failure) in patients with AMI.
Collapse
|
26
|
Sauler M, Bucala R, Lee PJ. Role of macrophage migration inhibitory factor in age-related lung disease. Am J Physiol Lung Cell Mol Physiol 2015; 309:L1-10. [PMID: 25957294 DOI: 10.1152/ajplung.00339.2014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 05/05/2015] [Indexed: 12/25/2022] Open
Abstract
The prevalence of many common respiratory disorders, including pneumonia, chronic obstructive lung disease, pulmonary fibrosis, and lung cancer, increases with age. Little is known of the host factors that may predispose individuals to such diseases. Macrophage migration inhibitory factor (MIF) is a potent upstream regulator of the immune system. MIF is encoded by variant alleles that occur commonly in the population. In addition to its role as a proinflammatory cytokine, a growing body of literature demonstrates that MIF influences diverse molecular processes important for the maintenance of cellular homeostasis and may influence the incidence or clinical manifestations of a variety of chronic lung diseases. This review highlights the biological properties of MIF and its implication in age-related lung disease.
Collapse
Affiliation(s)
- Maor Sauler
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut; and
| | - Richard Bucala
- Section of Rheumatology, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Patty J Lee
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut; and
| |
Collapse
|
27
|
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: 0.5] [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
|
28
|
Role of MIF in myocardial ischaemia and infarction: insight from recent clinical and experimental findings. Clin Sci (Lond) 2014; 127:149-61. [PMID: 24697297 DOI: 10.1042/cs20130828] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
First discovered in 1966 as an inflammatory cytokine, MIF (macrophage migration inhibitory factor) has been extensively studied for its pivotal role in a variety of inflammatory diseases, including rheumatoid arthritis and atherosclerosis. Although initial studies over a decade ago reported increases in circulating MIF levels following acute MI (myocardial infarction), the dynamic changes in MIF and its pathophysiological significance following MI have been unknown until recently. In the present review, we summarize recent experimental and clinical studies examining the diverse functions of MIF across the spectrum of acute MI from brief ischaemia to post-infarct healing. Following an acute ischaemic insult, MIF is rapidly released from jeopardized cardiomyocytes, followed by a persistent MIF production and release from activated immune cells, resulting in a sustained increase in circulating levels of MIF. Recent studies have documented two distinct actions of MIF following acute MI. In the supra-acute phase of ischaemia, MIF mediates cardioprotection via several distinct mechanisms, including metabolic activation, apoptosis suppression and antioxidative stress. In prolonged myocardial ischaemia, however, MIF promotes inflammatory responses with largely detrimental effects on cardiac function and remodelling. The pro-inflammatory properties of MIF are complex and involve MIF derived from cardiac and immune cells contributing sequentially to the innate immune response evoked by MI. Emerging evidence on the role of MIF in myocardial ischaemia and infarction highlights a significant potential for the clinical use of MIF agonists or antagonists and as a unique cardiac biomarker.
Collapse
|
29
|
Rassaf T, Weber C, Bernhagen J. Macrophage migration inhibitory factor in myocardial ischaemia/reperfusion injury. Cardiovasc Res 2014; 102:321-8. [PMID: 24675723 DOI: 10.1093/cvr/cvu071] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Acute myocardial infarction (AMI) remains one of the leading causes of death in the developed world. There is emerging evidence that the cytokine macrophage migration inhibitory factor (MIF) is a crucial player in AMI. Cardioprotection by MIF is likely to be a multifactorial phenomenon mediated by receptor-mediated signalling processes, intracellular protein-protein interactions, and enzymatic redox regulation. Co-ordinating several pathways in the ischaemic heart, MIF contributes to receptor-mediated regulation of cardioprotective AMP-activated protein kinase signalling, inhibition of pro-apoptotic cascades, and the reduction of oxidative stress in the post-ischaemic heart. Moreover, the cardioprotective properties of MIF are modulated by S-nitros(yl)ation. These effects in the pathophysiology of myocardial ischaemia/reperfusion injury qualify MIF as a promising therapeutic target in the future. We here summarize the findings of experimental and clinical studies and emphasize the therapeutic potential of MIF in AMI.
Collapse
Affiliation(s)
- Tienush Rassaf
- Medical Faculty, Division of Cardiology, Pulmonology and Vascular Medicine, University Hospital Düsseldorf, Moorenstrasse 5, Düsseldorf D-40225, Germany
| | | | | |
Collapse
|
30
|
Goetzenich A, Kraemer S, Rossaint R, Bleilevens C, Dollo F, Siry L, Rajabi-Alampour S, Beckers C, Soppert J, Lue H, Rex S, Bernhagen J, Stoppe C. The role of macrophage migration inhibitory factor in anesthetic-induced myocardial preconditioning. PLoS One 2014; 9:e92827. [PMID: 24667295 PMCID: PMC3965449 DOI: 10.1371/journal.pone.0092827] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 02/26/2014] [Indexed: 01/22/2023] Open
Abstract
Introduction Anesthetic-induced preconditioning (AIP) is known to elicit cardioprotective effects that are mediated at least in part by activation of the kinases AMPK and PKCε as well as by inhibition of JNK. Recent data demonstrated that the pleiotropic cytokine macrophage migration inhibitory factor (MIF) provides cardioprotection through activation and/or inhibition of kinases that are also known to mediate effects of AIP. Therefore, we hypothesized that MIF could play a key role in the AIP response. Methods Cardiomyocytes were isolated from rats and subjected to isoflurane preconditioning (4 h; 1.5 vol. %). Subsequently, MIF secretion and alterations in the activation levels of protective kinases were compared to a control group that was exposed to ambient air conditions. MIF secretion was quantified by ELISA and AIP-induced activation of protein kinases was assessed by Western blotting of cardiomyocyte lysates after isoflurane treatment. Results In cardiomyocytes, preconditioning with isoflurane resulted in a significantly elevated secretion of MIF that followed a biphasic behavior (30 min vs. baseline: p = 0.020; 24 h vs. baseline p = 0.000). Moreover, quantitative polymerase chain reaction demonstrated a significant increase in MIF mRNA expression 8 h after AIP. Of note, activation of AMPK and PKCε coincided with the observed peaks in MIF secretion and differed significantly from baseline. Conclusions These results suggest that the pleiotropic mediator MIF is involved in anesthetic-induced preconditioning of cardiomyocytes through stimulation of the protective kinases AMPK and PKCε.
Collapse
Affiliation(s)
- Andreas Goetzenich
- Department of Thoracic, Cardiac and Vascular Surgery, University Hospital, RWTH Aachen, Aachen, Germany
| | - Sandra Kraemer
- Department of Thoracic, Cardiac and Vascular Surgery, University Hospital, RWTH Aachen, Aachen, Germany
- Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Rolf Rossaint
- Department of Anesthesiology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Christian Bleilevens
- Department of Anesthesiology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Florian Dollo
- Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Laura Siry
- Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Setareh Rajabi-Alampour
- Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Christian Beckers
- Department of Thoracic, Cardiac and Vascular Surgery, University Hospital, RWTH Aachen, Aachen, Germany
- Department of Anesthesiology, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Josefin Soppert
- Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Hongqi Lue
- Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Steffen Rex
- Department of Anesthesiology and Cardiovascular Science, University Hospitals Gasthuisberg, KU Leuven, Leuven, Belgium
| | - Jürgen Bernhagen
- Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Aachen, Germany
- * E-mail: (JB); (CS)
| | - Christian Stoppe
- Department of Thoracic, Cardiac and Vascular Surgery, University Hospital, RWTH Aachen, Aachen, Germany
- Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University, Aachen, Germany
- * E-mail: (JB); (CS)
| |
Collapse
|
31
|
White DA, Su Y, Kanellakis P, Kiriazis H, Morand EF, Bucala R, Dart AM, Gao XM, Du XJ. Differential roles of cardiac and leukocyte derived macrophage migration inhibitory factor in inflammatory responses and cardiac remodelling post myocardial infarction. J Mol Cell Cardiol 2014; 69:32-42. [PMID: 24508700 DOI: 10.1016/j.yjmcc.2014.01.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 01/21/2014] [Accepted: 01/28/2014] [Indexed: 01/05/2023]
Abstract
Myocardial infarction (MI) provokes regional inflammation which facilitates the healing, whereas excessive inflammation leads to adverse cardiac remodelling. Our aim was to determine the role of macrophage migration inhibitory factor (MIF) in inflammation and cardiac remodelling following MI. Wild type (WT) or global MIF deficient (MIFKO) mice were subjected to coronary artery occlusion. Compared to WT mice, MIFKO mice had a significantly lower incidence of post-MI cardiac rupture (27% vs. 53%) and amelioration of cardiac remodelling. These were associated with suppressed myocardial leukocyte infiltration, inflammatory mediators' expression, and reduced activity of MMP-2, MMP-9, p38 and JNK MAPK. Infarct myocardium-derived or exogenous MIF mediated macrophage chemotaxis in vitro that was suppressed by inhibition of p38 MAPK or NF-κB. To further dissect the role of MIF derived from different cellular sources in post-MI cardiac remodelling, we generated chimeric mice with MIF deficiency either in bone marrow derived-cells (WT(KO)) or in somatic-cells (KO(WT)). Compared to WT and KO(WT) mice, WT(KO) mice had reduced rupture risk and ameliorated cardiac remodelling, associated with attenuated regional leukocyte infiltration and expression of inflammatory mediators. In contrast, KO(WT) mice had delayed healing and enhanced expression of M1 macrophage markers, but diminished expression of M2 markers during the early healing phase. In conclusion, global MIF deletion protects the heart from post-infarct cardiac rupture and remodelling through suppression of leukocyte infiltration and inflammation. Leukocyte-derived MIF promotes inflammatory responses after MI, whereas cardiac-derived MIF affects early but not ultimate healing process.
Collapse
Affiliation(s)
- David A White
- Baker IDI Heart and Diabetes Institute, Australia; Department of Cardiovascular Medicine, Alfred Hospital, Australia
| | - Yidan Su
- Baker IDI Heart and Diabetes Institute, Australia
| | | | | | - Eric F Morand
- Centre for Inflammatory Diseases, Southern Clinical School, Monash University, Australia
| | | | - Anthony M Dart
- Baker IDI Heart and Diabetes Institute, Australia; Department of Cardiovascular Medicine, Alfred Hospital, Australia; Department of Medicine, Central Clinical School, Monash University, Australia
| | - Xiao-Ming Gao
- Baker IDI Heart and Diabetes Institute, Australia; Department of Surgery, Central Clinical School, Monash University, Australia.
| | - Xiao-Jun Du
- Baker IDI Heart and Diabetes Institute, Australia; Department of Medicine, Central Clinical School, Monash University, Australia.
| |
Collapse
|
32
|
Ghigo A, Franco I, Morello F, Hirsch E. Myocyte signalling in leucocyte recruitment to the heart. Cardiovasc Res 2014; 102:270-80. [PMID: 24501328 DOI: 10.1093/cvr/cvu030] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Myocardial damage, by different noxious causes, triggers an inflammatory reaction driving post-injury repair mechanisms and chronic remodelling processes that are largely detrimental to cardiac function. Cardiomyocytes have recently emerged as key players in orchestrating this inflammatory response. Injured cardiomyocytes release damage-associated molecular pattern molecules, such as high-mobility group box 1 (HMGB1), DNA fragments, heat shock proteins, and matricellular proteins, which instruct surrounding healthy cadiomyocytes to produce inflammatory mediators. These mediators, mainly interleukin (IL)-1β, IL-6, macrophage chemoattractant protein (MCP)-1, and tumour necrosis factor α (TNF-α), in turn activate versatile signalling networks within surviving cardiomyocytes and trigger leucocyte activation and recruitment. In this review, we will focus on recently characterized signalling pathways activated in cardiomyocytes that mediate inflammatory responses during myocardial infarction, hypertensive heart disease, and myocarditis.
Collapse
Affiliation(s)
- Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, Torino, Italy
| | | | | | | |
Collapse
|
33
|
Chan W, White DA, Wang XY, Bai RF, Liu Y, Yu HY, Zhang YY, Fan F, Schneider HG, Duffy SJ, Taylor AJ, Du XJ, Gao W, Gao XM, Dart AM. Macrophage migration inhibitory factor for the early prediction of infarct size. J Am Heart Assoc 2013; 2:e000226. [PMID: 24096574 PMCID: PMC3835222 DOI: 10.1161/jaha.113.000226] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Early diagnosis and knowledge of infarct size is critical for the management of acute myocardial infarction (MI). We evaluated whether early elevated plasma level of macrophage migration inhibitory factor (MIF) is useful for these purposes in patients with ST-elevation MI (STEMI). METHODS AND RESULTS We first studied MIF level in plasma and the myocardium in mice and determined infarct size. MI for 15 or 60 minutes resulted in 2.5-fold increase over control values in plasma MIF levels while MIF content in the ischemic myocardium reduced by 50% and plasma MIF levels correlated with myocardium-at-risk and infarct size at both time-points (P < 0.01). In patients with STEMI, we obtained admission plasma samples and measured MIF, conventional troponins (TnI, TnT), high sensitive TnI (hsTnI), creatine kinase (CK), CK-MB, and myoglobin. Infarct size was assessed by cardiac magnetic resonance (CMR) imaging. Patients with chronic stable angina and healthy volunteers were studied as controls. Of 374 STEMI patients, 68% had elevated admission MIF levels above the highest value in healthy controls (> 41.6 ng/mL), a proportion similar to hsTnI (75%) and TnI (50%), but greater than other biomarkers studied (20% to 31%, all P < 0.05 versus MIF). Only admission MIF levels correlated with CMR-derived infarct size, ventricular volumes and ejection fraction (n = 42, r = 0.46 to 0.77, all P < 0.01) at 3 day and 3 months post-MI. CONCLUSION Plasma MIF levels are elevated in a high proportion of STEMI patients at the first obtainable sample and these levels are predictive of final infarct size and the extent of cardiac remodeling.
Collapse
Affiliation(s)
- William Chan
- Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
White DA, Fang L, Chan W, Morand EF, Kiriazis H, Duffy SJ, Taylor AJ, Dart AM, Du XJ, Gao XM. Pro-inflammatory action of MIF in acute myocardial infarction via activation of peripheral blood mononuclear cells. PLoS One 2013; 8:e76206. [PMID: 24098445 PMCID: PMC3788072 DOI: 10.1371/journal.pone.0076206] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Accepted: 08/21/2013] [Indexed: 12/11/2022] Open
Abstract
Objectives Macrophage migration inhibitory factor (MIF), a pro-inflammatory cytokine, has been implicated in the pathogenesis of multiple inflammatory disorders. We determined changes in circulating MIF levels, explored the cellular source of MIF, and studied the role of MIF in mediating inflammatory responses following acute myocardial infarction (MI). Methods and Results We recruited 15 patients with MI, 10 patients with stable angina and 10 healthy volunteers and measured temporal changes of MIF in plasma. Expression of MIF, matrix metalloproteinase-9 (MMP-9) and interleukin-6 (IL-6) in cultured peripheral blood mononuclear cells (PBMCs) and the media were measured by ELISA or real-time PCR. Compared to controls, plasma levels of MIF and IL-6 were significantly elevated at admission and 72 h post-MI. In contrast, expression of MIF, MMP-9 and IL-6 by PBMCs from MI patients was unchanged at admission, but significantly increased at 72 h. Addition of MIF activated cultured PBMCs by upregulating expression of inflammatory molecules and also synergistically enhanced stimulatory action of IL-1β which were inhibited by anti-MIF interventions. In a mouse MI model we observed similar changes in circulating MIF as seen in patients, with reciprocal significant increases in plasma MIF and reduction of MIF content in the infarct myocardium at 3 h after MI. MIF content in the infarct myocardium was restored at 72 h post-MI and was associated with robust macrophage infiltration. Further, anti-MIF intervention significantly reduced inflammatory cell infiltration and expression of monocyte chemoattractant protein-1 at 24 h and incidence of cardiac rupture in mice post-MI. Conclusion MI leads to a rapid release of MIF from the myocardium into circulation. Subsequently MIF facilitates PBMC production of pro-inflammatory mediators and myocardial inflammatory infiltration. Attenuation of these events, and post-MI cardiac rupture, by anti-MIF interventions suggests that MIF could be a potential therapeutic target following MI.
Collapse
Affiliation(s)
- David A. White
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Lu Fang
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - William Chan
- Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia
| | - Eric F. Morand
- Centre for Inflammatory Diseases, Monash University, Melbourne, Australia
| | - Helen Kiriazis
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Stephen J. Duffy
- Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Andrew J. Taylor
- Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Anthony M. Dart
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
- Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Xiao-Jun Du
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
- * E-mail: (X-MG); (X-JD)
| | - Xiao-Ming Gao
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
- Department of Surgery, Central Clinical School, Monash University, Melbourne, Australia
- * E-mail: (X-MG); (X-JD)
| |
Collapse
|
35
|
Rao F, Deng CY, Zhang QH, Xue YM, Xiao DZ, Kuang SJ, Lin QX, Shan ZX, Liu XY, Zhu JN, Yu XY, Wu SL. Involvement of Src tyrosine kinase and protein kinase C in the expression of macrophage migration inhibitory factor induced by H2O2 in HL-1 mouse cardiac muscle cells. Braz J Med Biol Res 2013; 46:746-51. [PMID: 24036910 PMCID: PMC3854426 DOI: 10.1590/1414-431x20132936] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Accepted: 06/06/2013] [Indexed: 11/22/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF), a pleiotropic cytokine, plays an
important role in the pathogenesis of atrial fibrillation; however, the upstream
regulation of MIF in atrial myocytes remains unclear. In the present study, we
investigated whether and how MIF is regulated in response to the
renin-angiotensin system and oxidative stress in atrium myocytes (HL-1 cells).
MIF protein and mRNA levels in HL-1 cells were assayed using immunofluorescence,
real-time PCR, and Western blot. The result indicated that MIF was expressed in
the cytoplasm of HL-1 cells. Hydrogen peroxide (H2O2), but
not angiotensin II, stimulated MIF expression in HL-1 cells.
H2O2-induced MIF protein and gene levels increased in
a dose-dependent manner and were completely abolished in the presence of
catalase. H2O2-induced MIF production was completely
inhibited by tyrosine kinase inhibitors genistein and PP1, as well as by protein
kinase C (PKC) inhibitor GF109203X, suggesting that redox-sensitive MIF
production is mediated through tyrosine kinase and PKC-dependent mechanisms in
HL-1 cells. These results suggest that MIF is upregulated by HL-1 cells in
response to redox stress, probably by the activation of Src and PKC.
Collapse
Affiliation(s)
- F Rao
- Guangdong Academy of Medical Sciences, Guangdong Cardiovascular Institute, Guangdong General Hospital, Department of Cardiology, Guangzhou, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Liehn EA, Kanzler I, Konschalla S, Kroh A, Simsekyilmaz S, Sönmez TT, Bucala R, Bernhagen J, Weber C. Compartmentalized protective and detrimental effects of endogenous macrophage migration-inhibitory factor mediated by CXCR2 in a mouse model of myocardial ischemia/reperfusion. Arterioscler Thromb Vasc Biol 2013; 33:2180-6. [PMID: 23868943 DOI: 10.1161/atvbaha.113.301633] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Here, we aimed to clarify the role of CXC chemokine receptor (CXCR) 2 in macrophage migration-inhibitory factor (MIF)-mediated effects after myocardial ischemia and reperfusion. As a pleiotropic chemokine-like cytokine, MIF has been identified to activate multiple receptors, including CD74 and CXCR2. In models of myocardial infarction, MIF exerts both proinflammatory effects and protective effects in cardiomyocytes. Similarly, CXCR2 displays opposing effects in resident versus circulating cells. APPROACH AND RESULTS Using bone marrow transplantation, we generated chimeric mice with Cxcr2(-/-) bone marrow-derived inflammatory cells and wild-type (wt) resident cells (wt/Cxcr2(-/-)), Cxcr2(-/-) cardiomyocytes and wt bone marrow-derived cells (Cxcr2(-/-)/wt), and wt controls reconstituted with wt bone marrow (wt/wt). All groups were treated with anti-MIF or isotype control antibody before they underwent myocardial ischemia and reperfusion. Blocking MIF increased infarction size and impaired cardiac function in wt/wt and wt/CXCR2(-/-) mice but ameliorated functional parameters in Cxcr2(-/-)/wt mice, as analyzed by echocardiography and Langendorff perfusion. Neutrophil infiltration and angiogenesis were unaltered by MIF blockade or Cxcr2 deficiency. Monocyte infiltration was blunted in wt/Cxcr2(-/-) mice and reduced by MIF blockade in wt/wt and Cxcr2(-/-)/wt mice. Furthermore, MIF blockade attenuated collagen content in all groups in a CXCR2-independent manner. CONCLUSIONS The compartmentalized and opposing effects of MIF after myocardial ischemia and reperfusion are largely mediated by CXCR2. Although MIF confers protective effects by improving myocardial healing and function through CXCR2 in resident cells, thereby complementing paracrine effects through CD74/AMP-activated protein kinase, it exerts detrimental effects on CXCR2-bearing inflammatory cells by increasing monocyte infiltration and impairing heart function. These dichotomous findings should be considered when developing novel therapeutic strategies to treat myocardial infarction.
Collapse
Affiliation(s)
- Elisa A Liehn
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
AMP-activated protein kinase (AMPK) is a stress-activated kinase that functions as a cellular fuel gauge and master metabolic regulator. Recent investigation has elucidated novel molecular mechanisms of AMPK regulation and important biological actions of the AMPK pathway that are highly relevant to cardiovascular disease. Activation of the intrinsic AMPK pathway plays an important role in the myocardial response to ischemia, pressure overload, and heart failure. Pharmacological activation of AMPK shows promise as a therapeutic strategy in the treatment of heart disease. The purpose of this review is to assess how recent discoveries have extended and in some cases challenged existing paradigms, providing new insights into the regulation of AMPK, its diverse biological actions, and therapeutic potential in the heart.
Collapse
Affiliation(s)
- Vlad G Zaha
- Department of Internal Medicine, Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | | |
Collapse
|
38
|
Koga K, Kenessey A, Ojamaa K. Macrophage migration inhibitory factor antagonizes pressure overload-induced cardiac hypertrophy. Am J Physiol Heart Circ Physiol 2012; 304:H282-93. [PMID: 23144312 DOI: 10.1152/ajpheart.00595.2012] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Macrophage migration inhibitory factor (MIF) functions as a proinflammatory cytokine when secreted from the cell, but it also exhibits antioxidant properties by virtue of its intrinsic oxidoreductase activity. Since increased production of ROS is implicated in the development of left ventricular hypertrophy, we hypothesized that the redox activity of MIF protects the myocardium when exposed to hemodynamic stress. In a mouse model of myocardial hypertrophy induced by transverse aortic coarctation (TAC) for 10 days, we showed that growth of the MIF-deficient heart was significantly greater by 32% compared with wild-type (WT) TAC hearts and that fibrosis was increased by fourfold (2.62 ± 0.2% vs. 0.6 ± 0.1%). Circulating MIF was increased in TAC animals, and expression of MIF receptor, CD74, was increased in the hypertrophic myocardium. Gene expression analysis showed a 10-fold increase (P < 0.01) in ROS-generating mitochondrial NADPH oxidase and 2- to 3-fold reductions (P < 0.01) in mitochondrial SOD2 and mitochondrial aconitase activities, indicating enhanced oxidative injury in the hypertrophied MIF-deficient ventricle. Hypertrophic signaling pathways showed that phosphorylation of cytosolic glycogen synthase kinase-3α was greater (P < 0.05) at baseline in MIF-deficient hearts than in WT hearts and remained elevated after 10-day TAC. In the hemodynamically stressed MIF-deficient heart, nuclear p21(CIP1) increased sevenfold (P < 0.01), and the cytosolic increase of phospho-p21(CIP1) was significantly greater than in WT TAC hearts. We conclude that MIF antagonizes myocardial hypertrophy and fibrosis in response to hemodynamic stress by maintaining a redox homeostatic phenotype and attenuating stress-induced activation of hypertrophic signaling pathways.
Collapse
Affiliation(s)
- Kiyokazu Koga
- Center for Heart and Lung Research, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | | | | |
Collapse
|
39
|
Brain miffed by macrophage migration inhibitory factor. Int J Cell Biol 2012; 2012:139573. [PMID: 22973314 PMCID: PMC3438795 DOI: 10.1155/2012/139573] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 07/06/2012] [Accepted: 07/12/2012] [Indexed: 12/31/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a cytokine which also exhibits enzymatic properties like oxidoreductase and tautomerase. MIF plays a pivotal role in innate and acquired immunity as well as in the neuroendocrine axis. Since it is involved in the pathogenesis of acute and chronic inflammation, neoangiogenesis, and cancer, MIF and its signaling components are considered suitable targets for therapeutic intervention in several fields of medicine. In neurodegenerative and neurooncological diseases, MIF is a highly relevant, but still a hardly investigated mediator. MIF operates via intracellular protein-protein interaction as well as in CD74/CXCR2/CXCR4 receptor-mediated pathways to regulate essential cellular systems such as redox balance, HIF-1, and p53-mediated senescence and apoptosis as well as multiple signaling pathways. Acting as an endogenous glucocorticoid antagonist, MIF thus represents a relevant resistance gene in brain tumor therapies. Alongside this dual action, a functional homolog-annotated D-dopachrome tautomerase/MIF-2 has been uncovered utilizing the same cell surface receptor signaling cascade as MIF. Here we review MIF actions with respect to redox regulation in apoptosis and in tumor growth as well as its extracellular function with a focus on its potential role in brain diseases. We consider the possibility of MIF targeting in neurodegenerative processes and brain tumors by novel MIF-neutralizing approaches.
Collapse
|
40
|
Ishii T, Warabi E, Yanagawa T. Novel roles of peroxiredoxins in inflammation, cancer and innate immunity. J Clin Biochem Nutr 2012; 50:91-105. [PMID: 22448089 PMCID: PMC3303482 DOI: 10.3164/jcbn.11-109] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 09/20/2011] [Indexed: 02/06/2023] Open
Abstract
Peroxiredoxins possess thioredoxin or glutathione peroxidase and chaperone-like activities and thereby protect cells from oxidative insults. Recent studies, however, reveal additional functions of peroxiredoxins in gene expression and inflammation-related biological reactions such as tissue repair, parasite infection and tumor progression. Notably, peroxiredoxin 1, the major mammalian peroxiredoxin family protein, directly interacts with transcription factors such as c-Myc and NF-κB in the nucleus. Additionally, peroxiredoxin 1 is secreted from some cells following stimulation with TGF-β and other cytokines and is thus present in plasma and body fluids. Peroxiredoxin 1 is now recognized as one of the pro-inflammatory factors interacting with toll-like receptor 4, which triggers NF-κB activation and other signaling pathways to evoke inflammatory reactions. Some cancer cells release peroxiredoxin 1 to stimulate toll-like receptor 4-mediated signaling for their progression. Interestingly, peroxiredoxins expressed in protozoa and helminth may modulate host immune responses partly through toll-like receptor 4 for their survival and progression in host. Extracellular peroxiredoxin 1 and peroxiredoxin 2 are known to enhance natural killer cell activity and suppress virus-replication in cells. Peroxiredoxin 1-deficient mice show reduced antioxidant activities but also exhibit restrained tissue inflammatory reactions under some patho-physiological conditions. Novel functions of peroxiredoxins in inflammation, cancer and innate immunity are the focus of this review.
Collapse
Affiliation(s)
- Tetsuro Ishii
- Majors of Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | | | | |
Collapse
|
41
|
Kimura H, Sato Y, Tajima Y, Suzuki H, Yukitake H, Imaeda T, Kajino M, Oki H, Takizawa M, Tanida S. BTZO-1, a cardioprotective agent, reveals that macrophage migration inhibitory factor regulates ARE-mediated gene expression. ACTA ACUST UNITED AC 2011; 17:1282-94. [PMID: 21168764 DOI: 10.1016/j.chembiol.2010.10.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2010] [Revised: 10/08/2010] [Accepted: 10/12/2010] [Indexed: 11/15/2022]
Abstract
In a screening program to discover therapeutic drugs for heart diseases, we identified BTZO-1, a 1,3-benzothiazin-4-one derivative, which activated antioxidant response element (ARE)-mediated gene expression and suppressed oxidative stress-induced cardiomyocyte apoptosis in vitro. An active BTZO-1 derivative for ARE-activation protected heart tissue during ischemia/reperfusion injury in rats. Macrophage migration inhibitory factor (MIF), which is known to protect cells from oxidative insult, was identified as a specific BTZO-1-binding protein. BTZO-1 binds to MIF with a K(d) of 68.6 nM, and its binding required the intact N-terminal Pro1. MIF, in the presence of BTZO-1, activated the glutathione S-transferase Ya subunit (GST Ya) gene ARE, whereas reduction of cellular MIF protein levels by siRNA suppressed BTZO-1-induced GST Ya expression. These results suggest that BTZO-1 activates the GST Ya gene ARE by interacting with MIF.
Collapse
Affiliation(s)
- Haruhide Kimura
- Pharmaceutical Research Division, Takeda Pharmaceutical Co. Ltd., 17-85, Jusohonmachi 2-chome, Yodogawa-ku, Osaka 532-8686, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Koga K, Kenessey A, Powell SR, Sison CP, Miller EJ, Ojamaa K. Macrophage migration inhibitory factor provides cardioprotection during ischemia/reperfusion by reducing oxidative stress. Antioxid Redox Signal 2011; 14:1191-202. [PMID: 20831446 DOI: 10.1089/ars.2010.3163] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Macrophage migration inhibitory factor (MIF) is a multifunctional protein that exhibits an intrinsic thiol protein oxidoreductase activity and proinflammatory activities. In the present study to examine intracellular MIF redox function, exposure of MIF-deficient cardiac fibroblasts to oxidizing conditions resulted in a 2.3-fold increase (p < 0.001) in intracellular ROS that could be significantly reduced by adenoviral-mediated reexpression of recombinant MIF. In an animal model of myocardial injury by ischemia/reperfusion (I/R), MIF-deficient hearts exhibited higher levels of oxidative stress than did wild-type hearts, as measured by significantly higher oxidized glutathione levels (decreased GSH/GSSG ratio), increased protein oxidation, reduced aconitase activity, and increased mitochondrial injury (increased cytochrome c release). The increased myocardial oxidative stress after I/R was reflected by larger infarct size (INF) in MIF-deficient hearts versus wild-type (WT) hearts (21 ± 6% vs. 8 ± 3% INF/LV; p < 0.05). In vivo hemodynamic measurements showed that left ventricular (LV) contractile function of MIF-deficient hearts subjected to 15-min ischemia failed to recover during reperfusion compared with WT hearts (LV developed pressure and ± dP/dt; p = 0.02). These data represent the first in vivo evidence in support of a cardioprotective role of MIF in the postischemic heart by reducing oxidative stress.
Collapse
Affiliation(s)
- Kiyokazu Koga
- Elmezzi Graduate School of Molecular Medicine, The Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, New York, USA
| | | | | | | | | | | |
Collapse
|
43
|
Yu XY, Chen HM, Liang JL, Lin QX, Tan HH, Fu YH, Liu XY, Shan ZX, Li XH, Yang HZ, Yang M, Li Y, Lin SG. Hyperglycemic myocardial damage is mediated by proinflammatory cytokine: macrophage migration inhibitory factor. PLoS One 2011; 6:e16239. [PMID: 21283592 PMCID: PMC3026813 DOI: 10.1371/journal.pone.0016239] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Accepted: 12/13/2010] [Indexed: 01/07/2023] Open
Abstract
Background Diabetes has been regarded as an inflammatory condition which is associated with left ventricular diastolic dysfunction (LVDD). The purpose of this study was to examine the expression levels of macrophage migration inhibitory factor (MIF) and G protein-coupled receptor kinase 2 (GRK2) in patients with early diabetic cardiomyopathy, and to investigate the mechanisms involved in MIF expression and GRK2 activation. Methods 83 patients in the age range of 30-64 years with type 2 diabetes and 30 matched healthy men were recruited. Left ventricular diastolic function was evaluated by cardiac Doppler echocardiography. Plasma MIF levels were determined by ELISA. To confirm the clinical observation, we also studied MIF expression in prediabetic rats with impaired glucose tolerance (IGT) and relationship between MIF and GRK2 expression in H9C2 cardiomyoblasts exposed to high glucose. Results Compared with healthy subjects, patients with diabetes have significantly increased levels of plasma MIF which was further increased in diabetic patients with Left ventricular diastolic dysfunction (LVDD). The increased plasma MIF levels in diabetic patients correlated with plasma glucose, glycosylated hemoglobin and urine albumin levels. We observed a significant number of TUNEL-positive cells in the myocardium of IGT-rats but not in the control rats. Moreover, we found higher MIF expression in the heart of IGT with cardiac dysfunction compared to that of the controls. In H9C2 cardiomyoblast cells, MIF and GRK2 expression was significantly increased in a glucose concentration-dependant manner. Furthermore, GRK2 expression was abolished by siRNA knockdown of MIF and by the inhibition of CXCR4 in H9C2 cells. Conclusions Our findings indicate that hyperglycemia is a causal factor for increased levels of pro-inflammatory cytokine MIF which plays a role in the development of cardiomyopathy occurring in patients with type 2 diabetes. The elevated levels of MIF are associated with cardiac dysfunction in diabetic patients, and the MIF effects are mediated by GRK2.
Collapse
Affiliation(s)
- Xi-Yong Yu
- Medical Research Center, Guangdong Provincial Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
- * E-mail: (X-YY); (YL); (S-GL)
| | - Hong-Mei Chen
- Medical Research Center, Guangdong Provincial Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
- Division of Endocrinology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Jia-Liang Liang
- Medical Research Center, Guangdong Provincial Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Qiu-Xiong Lin
- Medical Research Center, Guangdong Provincial Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Hong-Hong Tan
- Medical Research Center, Guangdong Provincial Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Yong-Heng Fu
- Medical Research Center, Guangdong Provincial Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Xiao-Ying Liu
- Medical Research Center, Guangdong Provincial Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Zhi-Xin Shan
- Medical Research Center, Guangdong Provincial Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Xiao-Hong Li
- Medical Research Center, Guangdong Provincial Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Hua-Zhang Yang
- Division of Endocrinology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Min Yang
- Medical Research Center, Guangdong Provincial Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Yangxin Li
- Texas Heart Institute and University of Texas Health Science Center, Houston, Texas, United States of America
- * E-mail: (X-YY); (YL); (S-GL)
| | - Shu-Guang Lin
- Medical Research Center, Guangdong Provincial Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
- * E-mail: (X-YY); (YL); (S-GL)
| |
Collapse
|
44
|
Liang JL, Xiao DZ, Liu XY, Lin QX, Shan ZX, Zhu JN, Lin SG, Yu XY. High glucose induces apoptosis in AC16 human cardiomyocytes via macrophage migration inhibitory factor and c-Jun N-terminal kinase. Clin Exp Pharmacol Physiol 2010; 37:969-73. [DOI: 10.1111/j.1440-1681.2010.05420.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
45
|
Involvement of Src in L-type Ca2+ channel depression induced by macrophage migration inhibitory factor in atrial myocytes. J Mol Cell Cardiol 2009; 47:586-94. [DOI: 10.1016/j.yjmcc.2009.08.030] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Revised: 08/24/2009] [Accepted: 08/29/2009] [Indexed: 11/22/2022]
|
46
|
Harrison RA, Sumners C. Redox regulation of macrophage migration inhibitory factor expression in rat neurons. Biochem Biophys Res Commun 2009; 390:171-5. [PMID: 19799867 DOI: 10.1016/j.bbrc.2009.09.112] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2009] [Accepted: 09/25/2009] [Indexed: 01/29/2023]
Abstract
Macrophage migration inhibitory factor (MIF) expression is induced by angiotensin II (Ang II) in normal rat neurons and serves a negative regulatory role by blunting the chronotropic actions of this peptide. The aim here was to determine whether hydrogen peroxide (H(2)O(2)), a reactive oxygen species (ROS) that is a key intracellular mediator of the neuronal actions of Ang II, is a trigger for MIF production in neurons. Thus, we tested the effects of H(2)O(2) on MIF expression in primary neuronal cultures from newborn normotensive (Wistar Kyoto [WKY] or Sprague-Dawley [SD]) rat brain, cells that respond to Ang II by increasing MIF levels. Treatment of WKY or SD rat neuronal cultures with a non-cytotoxic concentration of H(2)O(2) elicited a significant, time-dependent increase in MIF mRNA and protein levels. Glucose oxidase, which produces H(2)O(2) via oxidation of glucose in the cell-culture medium, elicited a similar increase in neuronal MIF mRNA levels. The stimulatory action of H(2)O(2) was not apparent in neuronal cultures from spontaneously hypertensive rats (SHR), cells that fail to express increased MIF in response to Ang II. Finally, preincubation of SD rat cultures with either polyethylene glycol-catalase or actinomycin D abolished the H(2)O(2)-induced increase in MIF, suggesting that this ROS is acting intracellularly to increase transcription of the MIF gene. These results suggest the presence of a redox regulatory mechanism for induction of MIF in normotensive rat neurons.
Collapse
Affiliation(s)
- Rachael A Harrison
- Department of Physiology and Functional Genomics & McKnight Brain Institute, University of Florida, Gainesville, FL 32610-0274, USA
| | | |
Collapse
|
47
|
Abstract
Over the past decade, AMP-activated protein kinase (AMPK) has emerged as an important intracellular signalling pathway in the heart. Activated AMPK stimulates the production of ATP by regulating key steps in both glucose and fatty acid metabolism. It has an inhibitory effect on cardiac protein synthesis. AMPK also interacts with additional intracellular signalling pathways in a coordinated network that modulates essential cellular processes in the heart. Evidence is accumulating that AMPK may protect the heart from ischaemic injury and limit the development of cardiac myocyte hypertrophy to various stimuli. Heart AMPK is activated by hormones, cytokines and oral hypoglycaemic drugs that are used in the treatment of type 2 diabetes. The tumour suppressor LKB1 is the major regulator of AMPK activity, but additional upstream kinases and protein phosphatases also contribute. Mutations in the regulatory gamma2 subunit of AMPK lead to an inherited syndrome of hypertrophic cardiomyopathy and ventricular pre-excitation, which appears to be due to intracellular glycogen accumulation. Future research promises to elucidate the molecular mechanisms responsible for AMPK activation, novel downstream AMPK targets, and the therapeutic potential of targeting AMPK for the prevention and treatment of myocardial ischaemia or cardiac hypertrophy.
Collapse
Affiliation(s)
- A S Kim
- Departments of Internal Medicine and Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | | | | |
Collapse
|
48
|
Jian Z, Li JB, Ma RY, Chen L, Zhong QJ, Wang XF, Wang W, Hong Y, Xiao YB. Increase of macrophage migration inhibitory factor (MIF) expression in cardiomyocytes during chronic hypoxia. Clin Chim Acta 2009; 405:132-8. [PMID: 19394321 DOI: 10.1016/j.cca.2009.04.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2009] [Revised: 04/21/2009] [Accepted: 04/21/2009] [Indexed: 01/12/2023]
Abstract
BACKGROUND Macrophage migration inhibitory factor (MIF) might play an important role in the myocardium during chronic hypoxia because MIF protects the heart during myocardial ischemia by activating 5'-adenosine monophosphate activated protein kinase (AMPK). METHODS We investigated 35 infants with cyanotic or acyanotic cardiac defects and H9c2 embryonic rat cardiomyocytes to examine the effect of chronic hypoxia on the expression of MIF in vivo and in vitro, respectively. RESULTS We found out an increase of endogenous cardiac MIF expression positively correlated with degree of hypoxia. Also, AMPK activation was elevated while MIF expression was increased in cells exposed to long periods of hypoxia in vitro. There was no significant difference in the growth ratio of cells cultivated in long periods of hypoxia and normoxia. CONCLUSIONS The expression of MIF is significantly increased in cardiomyocytes exposed to chronic hypoxia, and the activation of AMPK was increased accordingly.
Collapse
Affiliation(s)
- Zhao Jian
- Department of Cardiovascular Surgery, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Street, Chongqing 400037, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Cho ML, Moon YM, Heo YJ, Woo YJ, Ju JH, Park KS, Kim SI, Park SH, Kim HY, Min JK. NF-κB inhibition leads to increased synthesis and secretion of MIF in human CD4+ T cells. Immunol Lett 2009; 123:21-30. [DOI: 10.1016/j.imlet.2009.01.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2008] [Revised: 01/19/2009] [Accepted: 01/25/2009] [Indexed: 10/21/2022]
|
50
|
Rendon BE, Willer SS, Zundel W, Mitchell RA. Mechanisms of macrophage migration inhibitory factor (MIF)-dependent tumor microenvironmental adaptation. Exp Mol Pathol 2009; 86:180-5. [PMID: 19186177 DOI: 10.1016/j.yexmp.2009.01.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2008] [Indexed: 01/08/2023]
Abstract
Since its activity was first reported in the mid-1960s, macrophage migration inhibitory factor (MIF) has gone from a cytokine activity modulating monocyte motility to a pleiotropic regulator of a vast array of cellular and biological processes. Studies in recent years suggest that MIF contributes to malignant disease progression on several different levels. Both circulating and intracellular MIF protein levels are elevated in cancer patients and MIF expression reportedly correlates with stage, metastatic spread and disease-free survival. Additionally, MIF expression positively correlates with angiogenic growth factor expression, microvessel density and tumor-associated neovascularization. Not coincidentally, MIF has recently been shown to contribute to tumoral hypoxic adaptation by promoting hypoxia-induced HIF-1alpha stabilization. Intriguingly, hypoxia is a strong regulator of MIF expression and secretion, suggesting that hypoxia-induced MIF acts as an amplifying factor for both hypoxia and normoxia-associated angiogenic growth factor expression in human malignancies. Combined, these findings suggest that MIF overexpression contributes to tumoral hypoxic adaptation and, by extension, therapeutic responsiveness and disease prognosis. This review summarizes recent literature on the contributions of MIF to tumor-associated angiogenic growth factor expression, neovascularization and hypoxic adaptation. We also will review recent efforts aimed at identifying and employing small-molecule antagonists of MIF as a novel approach to cancer therapeutics.
Collapse
Affiliation(s)
- Beatriz E Rendon
- JG Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | | | | | | |
Collapse
|