151
|
Janssen H, Wagner CS, Demmer P, Callies S, Sölter G, Loghmani-khouzani H, Hu N, Schuett H, Tietge UJF, Warnecke G, Larmann J, Theilmeier G. Acute perioperative-stress-induced increase of atherosclerotic plaque volume and vulnerability to rupture in apolipoprotein-E-deficient mice is amenable to statin treatment and IL-6 inhibition. Dis Model Mech 2015; 8:1071-80. [PMID: 26092124 PMCID: PMC4582096 DOI: 10.1242/dmm.018713] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 06/09/2015] [Indexed: 01/15/2023] Open
Abstract
Myocardial infarction and stroke are frequent after surgical procedures and consume a considerable amount of benefit of surgical therapy. Perioperative stress, induced by surgery, is composed of hemodynamic and inflammatory reactions. The effects of perioperative stress on atherosclerotic plaques are ill-defined. Murine models to investigate the influence of perioperative stress on plaque stability and rupture are not available. We developed a model to investigate the influence of perioperative stress on plaque growth and stability by exposing apolipoprotein-E-deficient mice, fed a high cholesterol diet for 7 weeks, to a double hit consisting of 30 min of laparotomy combined with a substantial blood loss (approximately 20% of total blood volume; 400 µl). The innominate artery was harvested 72 h after the intervention. Control groups were sham and baseline controls. Interleukin-6 (IL-6) and serum amyloid A (SAA) plasma levels were determined. Plaque load, vascular smooth muscle cell (VSMC) and macrophage content were quantified. Plaque stability was assessed using the Stary score and frequency of signs of plaque rupture were assessed. High-dose atorvastatin (80 mg/kg body weight/day) was administered for 6 days starting 3 days prior to the double hit. A single dose of an IL-6-neutralizing antibody or the fusion protein gp130-Fc selectively targeting IL-6 trans-signaling was subcutaneously injected. IL-6 plasma levels increased, peaking at 6 h after the intervention. SAA levels peaked at 24 h (n=4, P<0.01). Plaque volume increased significantly with the double hit compared to sham (n=8, P<0.01). More plaques were scored as complex or bearing signs of rupture after the double hit compared to sham (n=5-8, P<0.05). Relative VSMC and macrophage content remained unchanged. IL-6-inhibition or atorvastatin, but not blocking of IL-6 trans-signaling, significantly decreased plaque volume and complexity (n=8, P<0.01). Using this model, researchers will be able to further investigate the pathophysiology of perioperative plaque stability, which can result in myocardial infarction, and, additionally, to test potential protective strategies. Summary: We developed a model to study the dynamics of atherosclerotic plaque growth and stability following surgery, and show that IL-6 inhibition and statins beneficially affect plaque volume and complexity.
Collapse
Affiliation(s)
- Henrike Janssen
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany Department of Anesthesiology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Christian S Wagner
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Philipp Demmer
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Simone Callies
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Gesine Sölter
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Houra Loghmani-khouzani
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Niandan Hu
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Harald Schuett
- Department of Cardiology, University Hospital Marburg, 35043 Marburg, Germany
| | - Uwe J F Tietge
- Department of Pediatrics, University of Groningen, UMCG, NL-9700 Groningen, The Netherlands
| | - Gregor Warnecke
- Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany German Centre for Lung Research (DZL), 30625 Hannover, Germany
| | - Jan Larmann
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany Department of Anesthesiology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Gregor Theilmeier
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany Faculty VI - Medicine and Health Sciences, Dept of Health Services Sciences, University of Oldenburg, 26129 Germany
| |
Collapse
|
152
|
Cytokines in atherosclerosis: Key players in all stages of disease and promising therapeutic targets. Cytokine Growth Factor Rev 2015; 26:673-85. [PMID: 26005197 PMCID: PMC4671520 DOI: 10.1016/j.cytogfr.2015.04.003] [Citation(s) in RCA: 347] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 04/27/2015] [Indexed: 02/07/2023]
Abstract
Atherosclerosis, a chronic inflammatory disorder of the arteries, is responsible for most deaths in westernized societies with numbers increasing at a marked rate in developing countries. The disease is initiated by the activation of the endothelium by various risk factors leading to chemokine-mediated recruitment of immune cells. The uptake of modified lipoproteins by macrophages along with defective cholesterol efflux gives rise to foam cells associated with the fatty streak in the early phase of the disease. As the disease progresses, complex fibrotic plaques are produced as a result of lysis of foam cells, migration and proliferation of vascular smooth muscle cells and continued inflammatory response. Such plaques are stabilized by the extracellular matrix produced by smooth muscle cells and destabilized by matrix metalloproteinase from macrophages. Rupture of unstable plaques and subsequent thrombosis leads to clinical complications such as myocardial infarction. Cytokines are involved in all stages of atherosclerosis and have a profound influence on the pathogenesis of this disease. This review will describe our current understanding of the roles of different cytokines in atherosclerosis together with therapeutic approaches aimed at manipulating their actions.
Collapse
|
153
|
Moreno Velásquez I, Golabkesh Z, Källberg H, Leander K, de Faire U, Gigante B. Circulating levels of interleukin 6 soluble receptor and its natural antagonist, sgp130, and the risk of myocardial infarction. Atherosclerosis 2015; 240:477-81. [PMID: 25910182 DOI: 10.1016/j.atherosclerosis.2015.04.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 04/11/2015] [Accepted: 04/13/2015] [Indexed: 10/23/2022]
Abstract
OBJECTIVE To investigate the association between circulating levels of the soluble interleukin 6 receptor (sIL6R) and the soluble gp130 (sgp130) with myocardial infarction (MI) and to explore the potential interaction between sIL6R and sgp130 in this association. METHODS Study population is the Stockholm Heart Epidemiology Program (SHEEP), a population-based case-control study. SIL6R (ng/mL) and sgp130 (ng/mL) levels were measured in serum samples from 682 and 664 MI cases and 1103 and 1062 controls, respectively. Odds ratios (with 95% CIs) for MI were calculated using unconditional logistic regression. We adjusted for age, sex, hospital catchment area (crude) and for hypertension, diabetes, hypercholesterolemia, body mass index and smoking (adjusted model). Synergy index (S) and attributable proportion (AP) were estimated as measures of biological interaction. RESULTS Elevated concentrations of sIL6R (>75th percentile value) were associated with an increased occurrence of MI (compared to ≤75th percentile), with an adjusted OR of 1.4 (95% CI, 1.1-1.8). Very high (>90th percentile value) levels of sgp130 were associated with a reduced occurrence of MI [OR 0.7 (95% CI, 0.5-0.9)] (adjusted). There was an indication of a possible interaction between high sIL6R and low sgp130 (adjusted S score 1.7, 95% CI = 0.5-6.1; AP 0.19, 95% CI = -0.2-0.5), suggesting that low sgp130 levels may synergize with high sIL6R levels to increase risk of MI. CONCLUSIONS sIL6R and sgp130 had opposing associations with MI. Indeed, circulating sgp130 levels may modify the association of elevated sIL6R levels with MI.
Collapse
Affiliation(s)
- Ilais Moreno Velásquez
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Sweden
| | - Zahra Golabkesh
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Sweden
| | - Henrik Källberg
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Sweden
| | - Karin Leander
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Sweden
| | - Ulf de Faire
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Sweden; Dept of Cardiology, Karolinska University Hospital, Sweden
| | - Bruna Gigante
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Sweden; Division of Cardiovascular Medicine, Dept of Clinical Sciences, Danderyd University Hospital Stockholm, Sweden.
| |
Collapse
|
154
|
Garbers C, Aparicio-Siegmund S, Rose-John S. The IL-6/gp130/STAT3 signaling axis: recent advances towards specific inhibition. Curr Opin Immunol 2015; 34:75-82. [PMID: 25749511 DOI: 10.1016/j.coi.2015.02.008] [Citation(s) in RCA: 322] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 02/12/2015] [Accepted: 02/15/2015] [Indexed: 12/12/2022]
Abstract
Interleukin-6 has long been recognized as a prototypic pro-inflammatory cytokine that is involved in the pathogenesis of all inflammatory diseases. Activation of the gp130 homodimer by IL-6 leads to the initiation of Jak/STAT signaling, a pathway that is often constitutively switched on in inflammatory malignancies. However, a plethora of studies in the last decade has convincingly shown that only signaling via the soluble IL-6R (trans-signaling) accounts for the deleterious effects of IL-6, whereas classic signaling via the membrane-bound receptor is essential for the regenerative and anti-bacterial effects of IL-6 (classic signaling). In this review, we highlight recent developments in the field of IL-6 research, and specifically focus on advances towards a safe and specific inhibition of IL-6 trans-signaling.
Collapse
Affiliation(s)
- Christoph Garbers
- Institute of Biochemistry, Kiel University, Olshausenstrasse 40, Kiel, Germany
| | | | - Stefan Rose-John
- Institute of Biochemistry, Kiel University, Olshausenstrasse 40, Kiel, Germany.
| |
Collapse
|
155
|
Parnell E, Palmer TM, Yarwood SJ. The future of EPAC-targeted therapies: agonism versus antagonism. Trends Pharmacol Sci 2015; 36:203-14. [PMID: 25744542 PMCID: PMC4392396 DOI: 10.1016/j.tips.2015.02.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 02/03/2015] [Accepted: 02/05/2015] [Indexed: 02/06/2023]
Abstract
Although tractable to drug development, targeting of cAMP signalling has side effects. Selectively targeting EPAC1 and EPAC2 cAMP sensor enzymes may limit some of these off-target effects. EPAC agonists could be used to treat vascular inflammation (EPAC1) or type 2 diabetes (EPAC2). EPAC1 and EPAC2 antagonists could be used to treat heart disease.
Pharmaceutical manipulation of cAMP levels exerts beneficial effects through the regulation of the exchange protein activated by cAMP (EPAC) and protein kinase A (PKA) signalling routes. Recent attention has turned to the specific regulation of EPAC isoforms (EPAC1 and EPAC2) as a more targeted approach to cAMP-based therapies. For example, EPAC2-selective agonists could promote insulin secretion from pancreatic β cells, whereas EPAC1-selective agonists may be useful in the treatment of vascular inflammation. By contrast, EPAC1 and EPAC2 antagonists could both be useful in the treatment of heart failure. Here we discuss whether the best way forward is to design EPAC-selective agonists or antagonists and the current strategies being used to develop isoform-selective, small-molecule regulators of EPAC1 and EPAC2 activity.
Collapse
Affiliation(s)
- Euan Parnell
- Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Timothy M Palmer
- School of Pharmacy, University of Bradford, Bradford BD7 1DP, UK
| | - Stephen J Yarwood
- Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK.
| |
Collapse
|
156
|
Kraakman MJ, Kammoun HL, Allen TL, Deswaerte V, Henstridge DC, Estevez E, Matthews VB, Neill B, White DA, Murphy AJ, Peijs L, Yang C, Risis S, Bruce CR, Du XJ, Bobik A, Lee-Young RS, Kingwell BA, Vasanthakumar A, Shi W, Kallies A, Lancaster GI, Rose-John S, Febbraio MA. Blocking IL-6 trans-signaling prevents high-fat diet-induced adipose tissue macrophage recruitment but does not improve insulin resistance. Cell Metab 2015; 21:403-16. [PMID: 25738456 DOI: 10.1016/j.cmet.2015.02.006] [Citation(s) in RCA: 192] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 12/21/2014] [Accepted: 02/06/2015] [Indexed: 01/01/2023]
Abstract
Interleukin-6 (IL-6) plays a paradoxical role in inflammation and metabolism. The pro-inflammatory effects of IL-6 are mediated via IL-6 "trans-signaling," a process where the soluble form of the IL-6 receptor (sIL-6R) binds IL-6 and activates signaling in inflammatory cells that express the gp130 but not the IL-6 receptor. Here we show that trans-signaling recruits macrophages into adipose tissue (ATM). Moreover, blocking trans-signaling with soluble gp130Fc protein prevents high-fat diet (HFD)-induced ATM accumulation, but does not improve insulin action. Importantly, however, blockade of IL-6 trans-signaling, unlike complete ablation of IL-6 signaling, does not exacerbate obesity-induced weight gain, liver steatosis, or insulin resistance. Our data identify the sIL-6R as a critical chemotactic signal for ATM recruitment and suggest that selectively blocking IL-6 trans-signaling may be a more favorable treatment option for inflammatory diseases, compared with current treatments that completely block the action of IL-6 and negatively impact upon metabolic homeostasis.
Collapse
Affiliation(s)
- Michael J Kraakman
- Cellular and Molecular Metabolism Laboratory, BakerIDI Heart & Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Helene L Kammoun
- Cellular and Molecular Metabolism Laboratory, BakerIDI Heart & Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Tamara L Allen
- Cellular and Molecular Metabolism Laboratory, BakerIDI Heart & Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Virginie Deswaerte
- Vascular Biology and Atherosclerosis Laboratory, BakerIDI Heart & Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Darren C Henstridge
- Cellular and Molecular Metabolism Laboratory, BakerIDI Heart & Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Emma Estevez
- Cellular and Molecular Metabolism Laboratory, BakerIDI Heart & Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Vance B Matthews
- Cellular and Molecular Metabolism Laboratory, BakerIDI Heart & Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Bronwyn Neill
- Cellular and Molecular Metabolism Laboratory, BakerIDI Heart & Diabetes Institute, Melbourne, VIC 3004, Australia
| | - David A White
- Experimental Cardiology Laboratory, BakerIDI Heart & Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Andrew J Murphy
- Haematopoiesis and Leukocyte Biology Laboratory, BakerIDI Heart & Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Lone Peijs
- Cellular and Molecular Metabolism Laboratory, BakerIDI Heart & Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Christine Yang
- Cellular and Molecular Metabolism Laboratory, BakerIDI Heart & Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Steve Risis
- Cellular and Molecular Metabolism Laboratory, BakerIDI Heart & Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Clinton R Bruce
- Cellular and Molecular Metabolism Laboratory, BakerIDI Heart & Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Xiao-Jun Du
- Experimental Cardiology Laboratory, BakerIDI Heart & Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Alex Bobik
- Vascular Biology and Atherosclerosis Laboratory, BakerIDI Heart & Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Robert S Lee-Young
- Cellular and Molecular Metabolism Laboratory, BakerIDI Heart & Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Bronwyn A Kingwell
- Metabolic and Vascular Physiology Laboratory, BakerIDI Heart & Diabetes Institute, Melbourne, VIC 3004, Australia
| | | | - Wei Shi
- Walter & Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Computing and Information Systems, University of Melbourne, Parkville, VIC 3010, Australia
| | - Axel Kallies
- Walter & Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Graeme I Lancaster
- Cellular and Molecular Metabolism Laboratory, BakerIDI Heart & Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Stefan Rose-John
- Department of Biochemistry, Christian-Albrechts-Universität zu Kiel, 24098 Kiel, Germany
| | - Mark A Febbraio
- Cellular and Molecular Metabolism Laboratory, BakerIDI Heart & Diabetes Institute, Melbourne, VIC 3004, Australia.
| |
Collapse
|
157
|
Ackers-Johnson M, Talasila A, Sage AP, Long X, Bot I, Morrell NW, Bennett MR, Miano JM, Sinha S. Myocardin regulates vascular smooth muscle cell inflammatory activation and disease. Arterioscler Thromb Vasc Biol 2015; 35:817-28. [PMID: 25614278 DOI: 10.1161/atvbaha.114.305218] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Atherosclerosis, the cause of 50% of deaths in westernized societies, is widely regarded as a chronic vascular inflammatory disease. Vascular smooth muscle cell (VSMC) inflammatory activation in response to local proinflammatory stimuli contributes to disease progression and is a pervasive feature in developing atherosclerotic plaques. Therefore, it is of considerable therapeutic importance to identify mechanisms that regulate the VSMC inflammatory response. APPROACH AND RESULTS We report that myocardin, a powerful myogenic transcriptional coactivator, negatively regulates VSMC inflammatory activation and vascular disease. Myocardin levels are reduced during atherosclerosis, in association with phenotypic switching of smooth muscle cells. Myocardin deficiency accelerates atherogenesis in hypercholesterolemic apolipoprotein E(-/-) mice. Conversely, increased myocardin expression potently abrogates the induction of an array of inflammatory cytokines, chemokines, and adhesion molecules in VSMCs. Expression of myocardin in VSMCs reduces lipid uptake, macrophage interaction, chemotaxis, and macrophage-endothelial tethering in vitro, and attenuates monocyte accumulation within developing lesions in vivo. These results demonstrate that endogenous levels of myocardin are a critical regulator of vessel inflammation. CONCLUSIONS We propose myocardin as a guardian of the contractile, noninflammatory VSMC phenotype, with loss of myocardin representing a critical permissive step in the process of phenotypic transition and inflammatory activation, at the onset of vascular disease.
Collapse
Affiliation(s)
- Matthew Ackers-Johnson
- From the Department of Medicine, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom (M.A.-J., A.T., A.P.S., N.W.M., M.R.B., S.S.); Department of Medicine, AAB Cardiovascular Research Institute, West Henrietta, NY (X.L., J.M.M.); and Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (I.B.)
| | - Amarnath Talasila
- From the Department of Medicine, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom (M.A.-J., A.T., A.P.S., N.W.M., M.R.B., S.S.); Department of Medicine, AAB Cardiovascular Research Institute, West Henrietta, NY (X.L., J.M.M.); and Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (I.B.)
| | - Andrew P Sage
- From the Department of Medicine, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom (M.A.-J., A.T., A.P.S., N.W.M., M.R.B., S.S.); Department of Medicine, AAB Cardiovascular Research Institute, West Henrietta, NY (X.L., J.M.M.); and Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (I.B.)
| | - Xiaochun Long
- From the Department of Medicine, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom (M.A.-J., A.T., A.P.S., N.W.M., M.R.B., S.S.); Department of Medicine, AAB Cardiovascular Research Institute, West Henrietta, NY (X.L., J.M.M.); and Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (I.B.)
| | - Ilze Bot
- From the Department of Medicine, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom (M.A.-J., A.T., A.P.S., N.W.M., M.R.B., S.S.); Department of Medicine, AAB Cardiovascular Research Institute, West Henrietta, NY (X.L., J.M.M.); and Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (I.B.)
| | - Nicholas W Morrell
- From the Department of Medicine, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom (M.A.-J., A.T., A.P.S., N.W.M., M.R.B., S.S.); Department of Medicine, AAB Cardiovascular Research Institute, West Henrietta, NY (X.L., J.M.M.); and Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (I.B.)
| | - Martin R Bennett
- From the Department of Medicine, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom (M.A.-J., A.T., A.P.S., N.W.M., M.R.B., S.S.); Department of Medicine, AAB Cardiovascular Research Institute, West Henrietta, NY (X.L., J.M.M.); and Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (I.B.)
| | - Joseph M Miano
- From the Department of Medicine, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom (M.A.-J., A.T., A.P.S., N.W.M., M.R.B., S.S.); Department of Medicine, AAB Cardiovascular Research Institute, West Henrietta, NY (X.L., J.M.M.); and Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (I.B.)
| | - Sanjay Sinha
- From the Department of Medicine, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom (M.A.-J., A.T., A.P.S., N.W.M., M.R.B., S.S.); Department of Medicine, AAB Cardiovascular Research Institute, West Henrietta, NY (X.L., J.M.M.); and Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands (I.B.).
| |
Collapse
|
158
|
Abstract
Both experimental and clinical evidence accumulated over the last couple of decades has linked inflammatory activation to the initiation and progression of chronic heart failure (HF). Circulating levels of inflammatory mediators are associated with cardiac function and inform risk prediction in patients, but the effect of anti-inflammatory therapy in HF remains uncertain. Interleukin (IL)-6 type cytokines are central to the inflammatory response, and convey their signals through the ubiquitously expressed glycoprotein (gp) 130 receptor subunit. IL-6-type/gp130 signaling therefore represents an inflammatory nexus, with inherent potential for disease modification. This review focuses on the current knowledge of IL-6/gp130 signaling in relation to HF, with a particular emphasis on the role of soluble gp130 (sgp130), a signaling pathway modulator. Biological aspects of sgp130 and IL-6 signaling are discussed, as are potential novel therapeutic approaches to modulate this central inflammatory signaling pathway.
Collapse
|
159
|
Abstract
Atherosclerosis is an inflammatory disease of the vessel wall characterized by activation of the innate immune system, with macrophages as the main players, as well as the adaptive immune system, characterized by a Th1-dominant immune response. Cytokines play a major role in the initiation and regulation of inflammation. In recent years, many studies have investigated the role of these molecules in experimental models of atherosclerosis. While some cytokines such as TNF or IFNγ clearly had atherogenic effects, others such as IL-10 were found to be atheroprotective. However, studies investigating the different cytokines in experimental atherosclerosis revealed that the cytokine system is complex with both disease stage-dependent and site-specific effects. In this review, we strive to provide an overview of the main cytokines involved in atherosclerosis and to shed light on their individual role during atherogenesis.
Collapse
Affiliation(s)
- Pascal J H Kusters
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Academic Medical Center, L01-146.1, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University (LMU), Munich, Germany.
| |
Collapse
|
160
|
Imbalanced gp130 signalling in ApoE-deficient mice protects against atherosclerosis. Atherosclerosis 2014; 238:321-8. [PMID: 25545330 DOI: 10.1016/j.atherosclerosis.2014.12.037] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 11/28/2014] [Accepted: 12/21/2014] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Interleukin (IL)-6 is a key modulator of the acute phase response (APR), and while both are implicated in atherosclerosis, the pathological role of specific IL-6 signalling cascades is ill-defined. Since IL-6 employs the cytokine receptor gp130 to primarily activate the STAT3 pathway, here we evaluate whether gp130-dependent STAT3 activation modulates atherosclerosis. METHODS High-fat diet-induced atherosclerosis was established in ApoE(-/-) mice crossed with gp130(F/F) knock-in mice displaying elevated gp130-dependent STAT3 activation and production of the APR protein, serum amyloid A (SAA). Also generated were gp130(F/F):Stat3(-/+):ApoE(-/-) mice displaying genetically-normalised STAT3 activation and SAA levels, and bone marrow chimeras involving ApoE(-/-) and gp130(F/F):ApoE(-/-) mice. At 10 weeks post high-fat diet, aortic atherosclerotic lesions, including the presence of CD68(+) macrophages, and plasma lipid and SAA profiles, were assessed. RESULTS Aortic plaque development and plasma triglyceride levels in gp130(F/F):ApoE(-/-) mice were significantly reduced (3-fold, P < 0.001) compared to ApoE(-/-) littermates. By contrast, in gp130(F/F):ApoE(-/-) mice, atherosclerotic plaques contained augmented CD68(+) macrophage infiltrates, and plasma SAA levels were elevated, compared to ApoE(-/-) mice. Atherosclerotic lesion development and plasma triglyceride levels in gp130(F/F):ApoE(-/-) and gp130(F/F):Stat3(-/+):ApoE(-/-) mice were comparable, despite a significant (P < 0.05) reduction in macrophage numbers in lesions, and also plasma SAA levels, in gp130(F/F):Stat3(-/+):ApoE(-/-) mice. Aortic plaque development and plasma triglyceride levels were comparable in ApoE(-/-) mice reconstituted with gp130(F/F):ApoE(-/-) (ApoE(F/F:ApoE)) or ApoE(-/-) (ApoE(ApoE)) bone marrow cells. CONCLUSIONS Deregulation of gp130/STAT3 signalling augments the APR and macrophage infiltration during atherosclerosis without impacting on the development of aortic plaques.
Collapse
|
161
|
Spoto B, Mattace-Raso F, Sijbrands E, Leonardis D, Testa A, Pisano A, Pizzini P, Cutrupi S, Parlongo RM, D'Arrigo G, Tripepi G, Mallamaci F, Zoccali C. Association of IL-6 and a functional polymorphism in the IL-6 gene with cardiovascular events in patients with CKD. Clin J Am Soc Nephrol 2014; 10:232-40. [PMID: 25492254 DOI: 10.2215/cjn.07000714] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND OBJECTIVES High serum IL-6 is a major risk factor for cardiovascular disease (CVD) in the general population. This cytokine is substantially increased in patients with CKD, but it is still unknown whether the link between IL-6 and CVD in CKD is causal in nature. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS In a cohort of 755 patients with stages 2-5 CKD, consecutively recruited from 22 nephrology units in southern Italy, this study assessed the relationship of serum IL-6 with history of CVD, as well as with incident cardiovascular (CV) events (mean follow up±SD, 31±10 months) and used the functional polymorphism (-174 G/C) in the promoter of the IL-6 gene to investigate whether the link between IL-6 and CV events is causal. RESULTS In adjusted analyses, serum IL-6 above the median value was associated with history of CVD (P<0.001) and predicted the incidence rate of CV events (hazard ratio, 1.66; 95% confidence interval [95% CI], 1.11 to 2.49; P=0.01). Patients homozygous for the risk allele (C) of the -174 G/C polymorphism had higher levels of IL-6 than did those with other genotypes (P=0.04). Homozygous CC patients more frequently had a history of CVD (odds ratio, 2.15; 95% CI, 1.15 to 4.00; P=0.02) as well as a 87% higher rate of incident CV events (hazard ratio, 1.87; 95% CI, 1.02 to 3.44; P=0.04) compared with other genotypes. CONCLUSIONS In patients with stages 2-5 CKD, high serum IL-6 is associated with history of CVD and predicts incident CV events. The parallel relationship with history of CVD and incident CV events of the -174 G/C polymorphism in the IL-6 gene suggests that IL-6 may be causally involved in the high CV risk in this population.
Collapse
Affiliation(s)
- Belinda Spoto
- National Research Council of Italy-Institute of Clinical Physiology & Institute of Biomedicine and Molecular Immunology & Nephrology, Dialysis and Transplantation Unit of Reggio, Calabria, Italy; and
| | - Francesco Mattace-Raso
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Eric Sijbrands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Daniela Leonardis
- National Research Council of Italy-Institute of Clinical Physiology & Institute of Biomedicine and Molecular Immunology & Nephrology, Dialysis and Transplantation Unit of Reggio, Calabria, Italy; and
| | - Alessandra Testa
- National Research Council of Italy-Institute of Clinical Physiology & Institute of Biomedicine and Molecular Immunology & Nephrology, Dialysis and Transplantation Unit of Reggio, Calabria, Italy; and
| | - Anna Pisano
- National Research Council of Italy-Institute of Clinical Physiology & Institute of Biomedicine and Molecular Immunology & Nephrology, Dialysis and Transplantation Unit of Reggio, Calabria, Italy; and
| | - Patrizia Pizzini
- National Research Council of Italy-Institute of Clinical Physiology & Institute of Biomedicine and Molecular Immunology & Nephrology, Dialysis and Transplantation Unit of Reggio, Calabria, Italy; and
| | - Sebastiano Cutrupi
- National Research Council of Italy-Institute of Clinical Physiology & Institute of Biomedicine and Molecular Immunology & Nephrology, Dialysis and Transplantation Unit of Reggio, Calabria, Italy; and
| | - Rosa M Parlongo
- National Research Council of Italy-Institute of Clinical Physiology & Institute of Biomedicine and Molecular Immunology & Nephrology, Dialysis and Transplantation Unit of Reggio, Calabria, Italy; and
| | - Graziella D'Arrigo
- National Research Council of Italy-Institute of Clinical Physiology & Institute of Biomedicine and Molecular Immunology & Nephrology, Dialysis and Transplantation Unit of Reggio, Calabria, Italy; and
| | - Giovanni Tripepi
- National Research Council of Italy-Institute of Clinical Physiology & Institute of Biomedicine and Molecular Immunology & Nephrology, Dialysis and Transplantation Unit of Reggio, Calabria, Italy; and
| | - Francesca Mallamaci
- National Research Council of Italy-Institute of Clinical Physiology & Institute of Biomedicine and Molecular Immunology & Nephrology, Dialysis and Transplantation Unit of Reggio, Calabria, Italy; and
| | - Carmine Zoccali
- National Research Council of Italy-Institute of Clinical Physiology & Institute of Biomedicine and Molecular Immunology & Nephrology, Dialysis and Transplantation Unit of Reggio, Calabria, Italy; and
| |
Collapse
|
162
|
Máchal J, Pávková-Goldbergová M, Hlinomaz O, Groch L, Vašků A. Patients with chronic three-vessel disease in a 15-year follow-up study: genetic and non-genetic predictors of survival. Medicine (Baltimore) 2014; 93:e278. [PMID: 25526459 PMCID: PMC4603099 DOI: 10.1097/md.0000000000000278] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Genetic and non-genetic predictors of 15-year survival in patients with chronic three-vessel disease (3VD) were investigated. Coronary angiography was performed on 810 subjects with symptoms of stable ischemic heart disease in 1998. The patients with 3VD were genotyped for 23 candidate polymorphisms covering the PPAR-RXR pathway, matrix metalloproteinase-2, renin-angiotensin-aldosterone system, endothelin-1, cytokine genes, MTHFR and APO E variants. Fifteen-year survival data were obtained from the national insurance registry. All data were available in the case of 150 patients with 3VD. Statistical analysis used stepwise Cox regression with dominant, recessive, or additive mode of genetic expression. Involved variables included age, sex, BMI, blood pressure, diabetes, ejection fraction, left main stenosis, previously diagnosed coronary stenosis, myocardial infarction in personal history, and coronary bypass along with polymorphisms pre-selected by log-rank tests. Out of the 23 polymorphisms, four were included in the model construction. SNP in the IL-6 gene rs1800795 (-174 G/C) has been found to be a significant predictor of survival. This SNP was in a linkage disequilibrium with rs1800797 (-597 G/A) in the same gene (D'=1.0), which was also found to constitute a significant predictor of survival when rs1800795 was not included in the model construction. Age, increased BMI, diabetes, low EF, and left main stenosis were also significant predictors in all models. Age, increased BMI, diabetes, low ejection fraction, left main stenosis, and genetic variation in the IL-6 promoter were established as significant independent risk factors for the survival of patients with three-vessel disease.
Collapse
Affiliation(s)
- Jan Máchal
- From the Department of Pathophysiology, Faculty of Medicine, Masaryk University Brno, Czech Republic (JM, MPG, AV); International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic (JM, LG, OH); and First Department of Internal Medicine - Cardioangiology, St. Anne's University Hospital, Brno, Czech Republic (OH, LG)
| | | | | | | | | |
Collapse
|
163
|
Mortensen MB, Kjolby M, Gunnersen S, Larsen JV, Palmfeldt J, Falk E, Nykjaer A, Bentzon JF. Targeting sortilin in immune cells reduces proinflammatory cytokines and atherosclerosis. J Clin Invest 2014; 124:5317-22. [PMID: 25401472 DOI: 10.1172/jci76002] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 10/16/2014] [Indexed: 12/21/2022] Open
Abstract
Genome-wide association studies have identified a link between genetic variation at the human chromosomal locus 1p13.3 and coronary artery disease. The gene encoding sortilin (SORT1) has been implicated as the causative gene within the locus, as sortilin regulates hepatic lipoprotein metabolism. Here we demonstrated that sortilin also directly affects atherogenesis, independent of its regulatory role in lipoprotein metabolism. In a mouse model of atherosclerosis, deletion of Sort1 did not alter plasma cholesterol levels, but reduced the development of both early and late atherosclerotic lesions. We determined that sortilin is a high-affinity receptor for the proinflammatory cytokines IL-6 and IFN-γ. Moreover, macrophages and Th1 cells (both of which mediate atherosclerotic plaque formation) lacking sortilin had reduced secretion of IL-6 and IFN-γ, but not of other measured cytokines. Transfer of sortilin-deficient BM into irradiated atherosclerotic mice reduced atherosclerosis and systemic markers of inflammation. Together, these data demonstrate that sortilin influences cytokine secretion and that targeting sortilin in immune cells attenuates inflammation and reduces atherosclerosis.
Collapse
|
164
|
Mangge H, Almer G, Stelzer I, Reininghaus E, Prassl R. Laboratory medicine for molecular imaging of atherosclerosis. Clin Chim Acta 2014; 437:19-24. [DOI: 10.1016/j.cca.2014.06.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 06/24/2014] [Accepted: 06/30/2014] [Indexed: 12/30/2022]
|
165
|
Affiliation(s)
- José J Fuster
- Molecular Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Kenneth Walsh
- Molecular Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
166
|
Garbers C, Scheller J. Interleukin-6 and interleukin-11: same same but different. Biol Chem 2014; 394:1145-61. [PMID: 23740659 DOI: 10.1515/hsz-2013-0166] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 05/30/2013] [Indexed: 11/15/2022]
Abstract
The pleiotropic physiological functions of interleukin (IL-)6 type cytokines range from embryonic development and tissue homoeostasis to neuronal development and T cell differentiation. In contrast, imbalance of the well-controlled cytokine signaling network leads to chronic inflammatory diseases and cancer. IL-6 and IL-11 both signal through a homodimer of the ubiquitously expressed β-receptor glycoprotein 130 (gp130). Specificity is gained through an individual IL-6/IL-11 α-receptor, which does not directly participate in signal transduction, although the initial cytokine binding event to the α-receptor leads to the final complex formation with the β-receptors. Both cytokines activate the same downstream signaling pathways, which are predominantly the mitogen-activated protein kinase (MAPK)-cascade and the Janus kinase/signal transducer and activator of transcription (Jak/STAT) pathway. However, recent studies have highlighted divergent roles of the two related cytokines. Here, we will discuss how the biochemical similarities are translated into unique and non-redundant functions of IL-6 and IL-11 in vivo and illustrate strategies for cytokine-specific therapeutic intervention.
Collapse
Affiliation(s)
- Christoph Garbers
- Medical Faculty, Institute of Biochemistry and Molecular Biology II, Heinrich-Heine University, Universit a tsstr. 1, D-40225 Dusseldorf, Germany
| | | |
Collapse
|
167
|
Wonnerth A, Katsaros KM, Krychtiuk KA, Speidl WS, Kaun C, Thaler K, Huber K, Wojta J, Maurer G, Seljeflot I, Arnesen H, Weiss TW. Glycoprotein 130 polymorphism predicts soluble glycoprotein 130 levels. Metabolism 2014; 63:647-53. [PMID: 24629561 DOI: 10.1016/j.metabol.2014.02.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 01/29/2014] [Accepted: 02/09/2014] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Interleukin-6 (IL-6) is a key cytokine in inflammatory diseases. It exerts its biological function via binding to a homodimer of its signal transducer glycoprotein 130 (gp130). Soluble gp130 (sgp130) is the natural inhibitor of IL-6 trans-signalling. The aim of this study was to test a possible influence of the gp130 genotype on sgp130 serum levels. MATERIAL AND METHODS In two separate populations, subjects were genotyped for the gp130 polymorphism G148C. Sgp130, IL-6 and soluble interleukin-6 receptor (sIL-6R) levels were measured. The OSLO population consisted of 546 male subjects at high risk for CAD. The VIENNA population consisted of 299 male subjects with angiographically proven CAD. RESULTS In the OSLO population, 124 (22.7%) subjects were hetero- or homozygote for the rare C allele. Individuals carrying the polymorphism had significantly higher levels of sgp130. In a multivariate linear regression model this association remained significant (adjusted p=0.001). In the VIENNA population, 48 (16.1%) subjects were hetero- or homozygote for the rare C allele. Consistent with the former study, sgp130 levels were significantly higher in carriers of the polymorphism compared to wildtype carriers (adjusted p=0.038). In the VIENNA population, sgp130 levels were significantly higher in diabetic patients. In the OSLO population, sgp130 was higher in patients with increased body mass index and in smokers (p<0.05). CONCLUSIONS Sgp130 serum levels are significantly higher in subjects carrying the gp130 polymorphism G148C compared to wildtype carriers. This finding proposes a possible genetical influence on sgp130 levels which may alter individual coping mechanisms in inflammatory diseases.
Collapse
Affiliation(s)
- Anna Wonnerth
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria.
| | - Katharina M Katsaros
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Cluster for Cardiovascular Research
| | | | - Walter S Speidl
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Christoph Kaun
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Cluster for Cardiovascular Research
| | - Kylie Thaler
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Kurt Huber
- Ludwig Boltzmann Cluster for Cardiovascular Research; Department of Cardiology and Emergency Medicine, Wilhelminenspital, Vienna, Austria
| | - Johann Wojta
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Cluster for Cardiovascular Research
| | - Gerald Maurer
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Ingebjorg Seljeflot
- Centre of Clinical Heart Research, Oslo University Hospital, Ulleval, Norway; Faculty of Medicine, University of Oslo, Norway
| | - Harald Arnesen
- Centre of Clinical Heart Research, Oslo University Hospital, Ulleval, Norway
| | - Thomas W Weiss
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria; Department of Cardiology and Emergency Medicine, Wilhelminenspital, Vienna, Austria
| |
Collapse
|
168
|
Rinne P, Silvola JMU, Hellberg S, Stahle M, Liljenback H, Salomaki H, Koskinen E, Nuutinen S, Saukko P, Knuuti J, Saraste A, Roivainen A, Savontaus E. Pharmacological Activation of the Melanocortin System Limits Plaque Inflammation and Ameliorates Vascular Dysfunction in Atherosclerotic Mice. Arterioscler Thromb Vasc Biol 2014; 34:1346-54. [DOI: 10.1161/atvbaha.113.302963] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
169
|
Callegari A, Coons ML, Ricks JL, Rosenfeld ME, Scatena M. Increased calcification in osteoprotegerin-deficient smooth muscle cells: Dependence on receptor activator of NF-κB ligand and interleukin 6. J Vasc Res 2014; 51:118-31. [PMID: 24642764 DOI: 10.1159/000358920] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 01/14/2014] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Vascular calcification is highly correlated with cardiovascular disease morbidity and mortality. Osteoprotegerin (OPG) is a secreted decoy receptor for receptor activator of NF-κB ligand (RANKL). Inactivation of OPG in apolipoprotein E-deficient (ApoE-/-) mice increases lesion size and calcification. The mechanism(s) by which OPG is atheroprotective and anticalcific have not been entirely determined. We investigated whether OPG-deficient vascular smooth muscle cells (VSMCs) are more susceptible to mineralization and whether RANKL mediates this process. RESULTS Lesion-free aortas from 12-week-old ApoE-/-OPG-/- mice had spotty calcification, an appearance of osteochondrogenic factors and a decrease of smooth muscle markers when compared to ApoE-/-OPG+/+ aortas. In osteogenic conditions, VSMCs isolated from ApoE-/-OPG-/- (KO-VSMC) mice deposited more calcium than VSMCs isolated from ApoE-/-OPG+/+ (WT-VSMC) mice. Gene expression and biochemical analysis indicated accelerated osteochondrogenic differentiation. Ablation of RANKL signaling in KO-VSMCs rescued the accelerated calcification. While WT-VSMCs did not respond to RANKL treatment, KO-VSMCs responded with enhanced calcification and the upregulation of osteochondrogenic genes. RANKL strongly induced interleukin 6 (IL-6), which partially mediated RANKL-dependent calcification and gene expression in KO-VSMCs. CONCLUSIONS OPG inhibits vascular calcification by regulating the procalcific effects of RANKL on VSMCs and is thus a possible target for therapeutic intervention.
Collapse
Affiliation(s)
- Andrea Callegari
- Department of Bioengineering, University of Washington, Seattle, Wash., USA
| | | | | | | | | |
Collapse
|
170
|
Interleukin-6 signal transduction and its role in hepatic lipid metabolic disorders. Cytokine 2014; 66:133-42. [PMID: 24491813 DOI: 10.1016/j.cyto.2013.12.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Revised: 12/14/2013] [Accepted: 12/30/2013] [Indexed: 01/12/2023]
Abstract
Hepatic lipid dysregulation can lead to spectrum of metabolic disease conditions including metabolic syndrome (MS), fatty liver and diabetes. Liver lipids are regulated by a complex set of extra-hepatic and intra-hepatic factors including cellular cross-talk with variety of cells, inducing various cytokines. Interleukin 6(IL-6) is a pleiotropic cytokine that exerts both pro-inflammatory and anti-inflammatory effects on hepatic system through either JNK/STAT or ERK/MAPK signaling. Although, IL-6 has shown to protect the liver from fat storage in both rodent and human models and various IL-6(-/-) studies have supported this notion yet a question remains over its deleterious pro-inflammatory effects on hepatocytes. IL-6 ability to produce reactive oxygen species (ROS) and subsequently disturb the hepatic lipid balance has created a conundrum. Furthermore, IL-6 has shown to behave differently under different disease states within hepatocytes and hence, modulating the hepatic lipids accordingly. This review deals with the role of IL-6 on hepatic lipid metabolism and analyzes various data presented on this topic.
Collapse
|
171
|
Scheller J, Garbers C, Rose-John S. Interleukin-6: from basic biology to selective blockade of pro-inflammatory activities. Semin Immunol 2013; 26:2-12. [PMID: 24325804 DOI: 10.1016/j.smim.2013.11.002] [Citation(s) in RCA: 241] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 11/13/2013] [Indexed: 12/16/2022]
Abstract
Cytokines receptors exist in membrane bound and soluble form. A soluble form of the human IL-6R is generated by limited proteolysis and alternative splicing. The complex of IL-6 and soluble IL-6R stimulates target cells not stimulated by IL-6 alone, since they do not express the membrane bound IL-6R. We have named this process trans-signaling. Soluble gp130 is the natural inhibitor of IL-6/soluble IL-6R complex responses. Recombinant soluble gp130 protein is a molecular tool to discriminate between gp130 responses via membrane bound and soluble IL-6R responses. Neutralizing monoclonal antibodies for global blockade of IL-6 signaling and the sgp130Fc protein for selective blockade of IL-6 trans-signaling have been used in several animal models of human diseases. Using the sgp130Fc protein or sgp130Fc transgenic mice we demonstrate in models of inflammatory bowel disease, peritonitis, rheumatoid arthritis, atherosclerosis pancreatitis, colon cancer, ovarian cancer and pancreatic cancer, that IL-6 trans-signaling via the soluble IL-6R is the crucial step in the development and the progression of the disease. Therefore, sgp130Fc is a novel therapeutic agent for the treatment of chronic inflammatory diseases and cancer and it undergoes phase I clinical trials as an anti-inflammatory drug since June 2013.
Collapse
Affiliation(s)
- Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Christoph Garbers
- Institute of Biochemistry, Christian-Albrechts-University, Olshausenstrasse 40, Kiel, Germany
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-University, Olshausenstrasse 40, Kiel, Germany.
| |
Collapse
|
172
|
Khedoe PPSJ, Wong MC, Wagenaar GTM, Plomp JJ, van Eck M, Havekes LM, Rensen PCN, Hiemstra PS, Berbée JFP. The effect of PPE-induced emphysema and chronic LPS-induced pulmonary inflammation on atherosclerosis development in APOE*3-LEIDEN mice. PLoS One 2013; 8:e80196. [PMID: 24303000 PMCID: PMC3841138 DOI: 10.1371/journal.pone.0080196] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 09/30/2013] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is characterized by pulmonary inflammation, airways obstruction and emphysema, and is a risk factor for cardiovascular disease (CVD). However, the contribution of these individual COPD components to this increased risk is unknown. Therefore, the aim of this study was to determine the contribution of emphysema in the presence or absence of pulmonary inflammation to the increased risk of CVD, using a mouse model for atherosclerosis. Because smoke is a known risk factor for both COPD and CVD, emphysema was induced by intratracheal instillation of porcine pancreatic elastase (PPE). METHODS Hyperlipidemic APOE*3-Leiden mice were intratracheally instilled with vehicle, 15 or 30 µg PPE and after 4 weeks, mice received a Western-type diet (WTD). To study the effect of emphysema combined with pulmonary inflammation on atherosclerosis, mice received 30 µg PPE and during WTD feeding, mice were intranasally instilled with vehicle or low-dose lipopolysaccharide (LPS; 1 µg/mouse, twice weekly). After 20 weeks WTD, mice were sacrificed and emphysema, pulmonary inflammation and atherosclerosis were analysed. RESULTS Intratracheal PPE administration resulted in a dose-dependent increase in emphysema, whereas atherosclerotic lesion area was not affected by PPE treatment. Additional low-dose intranasal LPS administration induced a low-grade systemic IL-6 response, as compared to vehicle. Combining intratracheal PPE with intranasal LPS instillation significantly increased the number of pulmonary macrophages and neutrophils. Plasma lipids during the study were not different. LPS instillation caused a limited, but significant increase in the atherosclerotic lesion area. This increase was not further enhanced by PPE. CONCLUSION This study shows for the first time that PPE-induced emphysema both in the presence and absence of pulmonary inflammation does not affect atherosclerotic lesion development.
Collapse
Affiliation(s)
- P. Padmini S. J Khedoe
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
- * E-mail:
| | - Man C. Wong
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Gerry T. M. Wagenaar
- Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Jaap J. Plomp
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Miranda van Eck
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, Leiden, The Netherlands
| | - Louis M. Havekes
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, The Netherlands
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Netherlands Organization for Applied Scientific Research, Metabolic Health Research, Gaubius Laboratory, Leiden, The Netherlands
| | - Patrick C. N. Rensen
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Pieter S. Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jimmy F. P. Berbée
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
173
|
Flavanoids induce expression of the suppressor of cytokine signalling 3 (SOCS3) gene and suppress IL-6-activated signal transducer and activator of transcription 3 (STAT3) activation in vascular endothelial cells. Biochem J 2013; 454:283-93. [PMID: 23782265 PMCID: PMC3749869 DOI: 10.1042/bj20130481] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The atherogenic cytokine IL-6 (interleukin-6) induces pro-inflammatory gene expression in VECs (vascular endothelial cells) by activating the JAK (Janus kinase)/STAT3 (signal transducer and activator of transcription 3) signalling pathway, which is normally down-regulated by the STAT3-dependent induction of the E3 ubiquitin ligase component SOCS3 (suppressor of cytokine signalling 3). Novel treatments based on the regulation of SOCS3 protein levels could therefore have value in the treatment of diseases with an inflammatory component, such as atherosclerosis. To this end we carried out a screen of 1031 existing medicinal compounds to identify inducers of SOCS3 gene expression and identified the flavanoids naringenin and flavone as effective inducers of SOCS3 protein, mRNA and promoter activity. This was in contrast with the action of traditional JAK/STAT3 inhibitors and the polyphenol resveratrol, which effectively suppress SOCS3 gene expression. Both naringenin and flavone also effectively suppressed IL-6-stimulated phosphorylation of STAT3 (Tyr705) which led to suppression of IL-6-induction of the atherogenic STAT3 target gene MCP1 (monocyte chemotactic protein-1), suggesting that their ability to induce SOCS3 gene expression is STAT3-independent. Supporting this idea was the observation that the general kinase inhibitor compound C inhibits flavone- and cAMP-dependent, but not JAK-dependent, SOCS3 induction in VECs. Indeed, the ability of flavanoids to induce SOCS3 expression requires activation of the ERK (extracellular-signal-regulated kinase)-dependent transcription factor SP3, and not STAT3. In the present paper we therefore describe novel molecular actions of flavanoids, which control SOCS3 gene induction and suppression of STAT3 signalling in VECs. These mechanisms could potentially be exploited to develop novel anti-atherogenic therapies.
Collapse
|
174
|
Abstract
At least 468 individual genes have been manipulated by molecular methods to study their effects on the initiation, promotion, and progression of atherosclerosis. Most clinicians and many investigators, even in related disciplines, find many of these genes and the related pathways entirely foreign. Medical schools generally do not attempt to incorporate the relevant molecular biology into their curriculum. A number of key signaling pathways are highly relevant to atherogenesis and are presented to provide a context for the gene manipulations summarized herein. The pathways include the following: the insulin receptor (and other receptor tyrosine kinases); Ras and MAPK activation; TNF-α and related family members leading to activation of NF-κB; effects of reactive oxygen species (ROS) on signaling; endothelial adaptations to flow including G protein-coupled receptor (GPCR) and integrin-related signaling; activation of endothelial and other cells by modified lipoproteins; purinergic signaling; control of leukocyte adhesion to endothelium, migration, and further activation; foam cell formation; and macrophage and vascular smooth muscle cell signaling related to proliferation, efferocytosis, and apoptosis. This review is intended primarily as an introduction to these key signaling pathways. They have become the focus of modern atherosclerosis research and will undoubtedly provide a rich resource for future innovation toward intervention and prevention of the number one cause of death in the modern world.
Collapse
Affiliation(s)
- Paul N Hopkins
- Cardiovascular Genetics, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.
| |
Collapse
|
175
|
Kraakman MJ, Allen TL, Whitham M, Iliades P, Kammoun HL, Estevez E, Lancaster GI, Febbraio MA. Targeting gp130 to prevent inflammation and promote insulin action. Diabetes Obes Metab 2013; 15 Suppl 3:170-5. [PMID: 24003934 DOI: 10.1111/dom.12170] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 05/14/2013] [Indexed: 02/06/2023]
Abstract
Obesity and type 2 diabetes are now the most prevalent metabolic diseases in the Western world and the development of new strategies to treat these metabolic diseases is most warranted. Obesity results in a state of chronic low-grade inflammation in metabolically active tissues such as the liver, adipose tissue, brain and skeletal muscle. Work in our laboratory has focussed on the role of the cytokine interleukin-6 (IL)-6 and other IL-6-like cytokines that signal through the gp130 receptor complex. We have focussed on the role of blocking IL-6 trans-signalling to prevent inflammation on the one hand, and activating membrane-bound signalling to promote insulin sensitivity on the other hand. Since the cloning of the IL-6 gene nearly 30 years ago, a pattern has emerged associating IL-6 with a number of diseases associated with inflammation including rheumatoid arthritis (RA), Crohn's disease and several cancers. Accordingly, tocilizumab, an IL-6 receptor-inhibiting monoclonal antibody, is now useful for the treatment of RA. However, this may not be the most optimal strategy to block inflammation associated with IL-6 and may result in unwanted side effects that, paradoxically, could actually promote metabolic disease.
Collapse
Affiliation(s)
- M J Kraakman
- Cellular & Molecular Metabolism Laboratory, Baker IDI Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | |
Collapse
|
176
|
Alman AC, Kinney GL, Tracy RP, Maahs DM, Hokanson JE, Rewers MJ, Snell-Bergeon JK. Prospective association between inflammatory markers and progression of coronary artery calcification in adults with and without type 1 diabetes. Diabetes Care 2013; 36:1967-73. [PMID: 23340891 PMCID: PMC3687315 DOI: 10.2337/dc12-1874] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE The role of inflammation in the increased risk of cardiovascular disease in type 1 diabetes is unclear. We examined the association of inflammation and progression of coronary artery calcification (CAC)-a marker of subclinical atherosclerosis-in adults with and without type 1 diabetes. RESEARCH DESIGN AND METHODS A nested case-control study was performed within the prospective cohort of the Coronary Artery Calcification in Type 1 Diabetes (CACTI) study. Participants underwent two CAC measurements ~2.5 years apart. Case subjects (n = 204) were those with significant progression of CAC. Control subjects (n = 258) were frequency-matched to case subjects on diabetes status, sex, age, and baseline CAC status. Inflammatory marker assessments were performed on stored blood samples from baseline. A principal components analysis (PCA) was performed and a composite score derived from that analysis. The composite score was constructed by assigning a value of 1 for each PCA component where at least one of the markers exceeded the 75th percentile (range 0-4). Conditional logistic regression was used for the matching strategy. RESULTS The first two components of the PCA were modestly (odds ratio 1.38 [95% CI 1.08-1.77] and 1.27 [1.02-1.59], respectively) associated with CAC progression after adjustment for other risk factors. The composite score was more strongly associated with CAC progression for those with elevated markers in three or four of the principal components compared with those with none. CONCLUSIONS Measures of inflammation were associated with progression of CAC in a population of adults with and without type 1 diabetes.
Collapse
Affiliation(s)
- Amy C Alman
- Department of Epidemiology and Biostatistics, College of Public Health, University of South Florida, Tampa, Florida, USA.
| | | | | | | | | | | | | |
Collapse
|
177
|
van Diepen JA, Berbée JF, Havekes LM, Rensen PC. Interactions between inflammation and lipid metabolism: Relevance for efficacy of anti-inflammatory drugs in the treatment of atherosclerosis. Atherosclerosis 2013; 228:306-15. [PMID: 23518178 DOI: 10.1016/j.atherosclerosis.2013.02.028] [Citation(s) in RCA: 207] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2012] [Revised: 01/31/2013] [Accepted: 02/21/2013] [Indexed: 12/14/2022]
|
178
|
Drechsler M, Soehnlein O. The complexity of arterial classical monocyte recruitment. J Innate Immun 2013; 5:358-66. [PMID: 23571485 PMCID: PMC6741506 DOI: 10.1159/000348795] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 02/09/2012] [Accepted: 02/09/2012] [Indexed: 12/24/2022] Open
Abstract
Accumulation of classical monocytes is imperative for the progression of atherosclerosis. Hence, therapeutic interference with mechanisms of lesional monocyte recruitment, the primary mechanism controlling macrophage accumulation, may allow for targeting atheroprogression and its clinical complications. Here, we review the important role of classical monocytes in atheroprogression as well as their routes of arterial recruitment. We specifically highlight the role of cell adhesion molecules as well as of platelet-derived chemokines and neutrophil-borne alarmins.
Collapse
Affiliation(s)
- Maik Drechsler
- Institute for Cardiovascular Prevention, Ludwig-Maximilians University, Munich, Germany.
| | | |
Collapse
|
179
|
Zhang H, Taylor WR, Joseph G, Caracciolo V, Gonzales DM, Sidell N, Seli E, Blackshear PJ, Kallen CB. mRNA-binding protein ZFP36 is expressed in atherosclerotic lesions and reduces inflammation in aortic endothelial cells. Arterioscler Thromb Vasc Biol 2013; 33:1212-20. [PMID: 23559629 DOI: 10.1161/atvbaha.113.301496] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE We studied the expression and function of an mRNA-binding protein, zinc finger protein-36 (ZFP36), in vascular endothelial cells in vivo and in vitro. We tested the hypotheses that ZFP36 regulates inflammation in vascular endothelial cells and that it functions through direct binding to target cytokine mRNAs. We also tested whether ZFP36 inhibits nuclear factor-κB-mediated transcriptional responses in vascular endothelial cells. APPROACH AND RESULTS ZFP36 was minimally expressed in healthy aorta but was expressed in endothelial cells overlying atherosclerotic lesions in mice and humans. The protein was also expressed in macrophage foam cells of atherosclerosis. ZFP36 was expressed in human aortic endothelial cells in response to bacterial lipopolysaccharide, glucocorticoid, and forskolin, but not oxidized low-density lipoproteins or angiotensin II. Functional studies demonstrated that ZFP36 reduces the expression of inflammatory cytokines in target cells by 2 distinct mechanisms: ZFP36 inhibits nuclear factor-κB transcriptional activation and also binds to cytokine mRNAs, leading to reduced transcript stability. CONCLUSIONS ZFP36 is expressed in vascular endothelial cells and macrophage foam cells where it inhibits the expression of proinflammatory mRNA transcripts. The anti-inflammatory effects of ZFP36 in endothelial cells occur via both transcriptional and posttranscriptional mechanisms. Our data suggest that enhancing vascular ZFP36 expression might reduce vascular inflammation.
Collapse
Affiliation(s)
- Huanchun Zhang
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
180
|
Askevold ET, Nymo S, Ueland T, Gravning J, Wergeland R, Kjekshus J, Yndestad A, Cleland JG, McMurray JJ, Aukrust P, Gullestad L. Soluble Glycoprotein 130 Predicts Fatal Outcomes in Chronic Heart Failure. Circ Heart Fail 2013; 6:91-8. [DOI: 10.1161/circheartfailure.112.972653] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
Glycoprotein 130 (gp130) is the common signal-transducing receptor subunit of the interleukin-6 (IL-6) family, which may be involved in the progression of heart failure (HF). We hypothesized that soluble gp130 would provide prognostic information beyond that of IL-6 in a population with HF from the Controlled Rosuvastatin Multinational Trial in Heart Failure (CORONA).
Methods and Results—
The associations of soluble gp130 and IL-6 with morbidity, mortality, and mode of death were assessed by immunoassays in a subset of 1452 patients enrolled in the CORONA trial, which included patients with HF, aged ≥60 years, in New York Heart Association classes II to IV, who had ischemic heart disease and a reduced left ventricular ejection fraction. In multivariable analyses, including C-reactive protein, IL-6, troponin T, and N-terminal pro-B-type natriuretic peptide, elevated soluble gp130 (fifth quintile versus all lower quintiles) was associated with all-cause mortality (hazard ratio, 1.47 [1.11–1.93];
P
=0.006), cardiovascular mortality (hazard ratio, 1.38 [1.01–1.87];
P
=0.042), and death from worsening HF (hazard ratio, 1.85 [1.09–3.14];
P
=0.002), but not with the primary end point (composite of death from cardiovascular causes, nonfatal myocardial infarction, and nonfatal stroke; hazard ratio, 1.12 [0.84–1.50];
P
=0.44). Plasma IL-6 was not associated with outcomes in multivariable analyses.
Conclusions—
Marked elevations in soluble gp130 are associated with total and cardiovascular mortality, as well as deaths from worsening HF, in elderly patients with HF of ischemic cause
Clinical Trial Registration—
URL:
http://www.clinicaltrials.gov
. Unique identifier: NCT00206310.
Collapse
Affiliation(s)
- Erik Tandberg Askevold
- From the Department of Cardiology (E.T.A., J.K., L.G.), Research Institute of Internal Medicine (E.T.A., S.N., T.U., A.Y., P.A.), Section of Clinical Immunology and Infectious Diseases (P.A.), Institute for Surgical Research (J.G.), Department of Medical Biochemistry, Oslo University Hospital Rikshospitalet (R.W.), Faculty of Medicine (T.U., J.K., A.Y., P.A., L.G.), and K.G. Jebsen Cardiac Research Centre and Center for Heart Failure Research (E.T.A., S.N., J.G., J.K., A.Y., L.G.), University of
| | - Ståle Nymo
- From the Department of Cardiology (E.T.A., J.K., L.G.), Research Institute of Internal Medicine (E.T.A., S.N., T.U., A.Y., P.A.), Section of Clinical Immunology and Infectious Diseases (P.A.), Institute for Surgical Research (J.G.), Department of Medical Biochemistry, Oslo University Hospital Rikshospitalet (R.W.), Faculty of Medicine (T.U., J.K., A.Y., P.A., L.G.), and K.G. Jebsen Cardiac Research Centre and Center for Heart Failure Research (E.T.A., S.N., J.G., J.K., A.Y., L.G.), University of
| | - Thor Ueland
- From the Department of Cardiology (E.T.A., J.K., L.G.), Research Institute of Internal Medicine (E.T.A., S.N., T.U., A.Y., P.A.), Section of Clinical Immunology and Infectious Diseases (P.A.), Institute for Surgical Research (J.G.), Department of Medical Biochemistry, Oslo University Hospital Rikshospitalet (R.W.), Faculty of Medicine (T.U., J.K., A.Y., P.A., L.G.), and K.G. Jebsen Cardiac Research Centre and Center for Heart Failure Research (E.T.A., S.N., J.G., J.K., A.Y., L.G.), University of
| | - Jørgen Gravning
- From the Department of Cardiology (E.T.A., J.K., L.G.), Research Institute of Internal Medicine (E.T.A., S.N., T.U., A.Y., P.A.), Section of Clinical Immunology and Infectious Diseases (P.A.), Institute for Surgical Research (J.G.), Department of Medical Biochemistry, Oslo University Hospital Rikshospitalet (R.W.), Faculty of Medicine (T.U., J.K., A.Y., P.A., L.G.), and K.G. Jebsen Cardiac Research Centre and Center for Heart Failure Research (E.T.A., S.N., J.G., J.K., A.Y., L.G.), University of
| | - Ragnhild Wergeland
- From the Department of Cardiology (E.T.A., J.K., L.G.), Research Institute of Internal Medicine (E.T.A., S.N., T.U., A.Y., P.A.), Section of Clinical Immunology and Infectious Diseases (P.A.), Institute for Surgical Research (J.G.), Department of Medical Biochemistry, Oslo University Hospital Rikshospitalet (R.W.), Faculty of Medicine (T.U., J.K., A.Y., P.A., L.G.), and K.G. Jebsen Cardiac Research Centre and Center for Heart Failure Research (E.T.A., S.N., J.G., J.K., A.Y., L.G.), University of
| | - John Kjekshus
- From the Department of Cardiology (E.T.A., J.K., L.G.), Research Institute of Internal Medicine (E.T.A., S.N., T.U., A.Y., P.A.), Section of Clinical Immunology and Infectious Diseases (P.A.), Institute for Surgical Research (J.G.), Department of Medical Biochemistry, Oslo University Hospital Rikshospitalet (R.W.), Faculty of Medicine (T.U., J.K., A.Y., P.A., L.G.), and K.G. Jebsen Cardiac Research Centre and Center for Heart Failure Research (E.T.A., S.N., J.G., J.K., A.Y., L.G.), University of
| | - Arne Yndestad
- From the Department of Cardiology (E.T.A., J.K., L.G.), Research Institute of Internal Medicine (E.T.A., S.N., T.U., A.Y., P.A.), Section of Clinical Immunology and Infectious Diseases (P.A.), Institute for Surgical Research (J.G.), Department of Medical Biochemistry, Oslo University Hospital Rikshospitalet (R.W.), Faculty of Medicine (T.U., J.K., A.Y., P.A., L.G.), and K.G. Jebsen Cardiac Research Centre and Center for Heart Failure Research (E.T.A., S.N., J.G., J.K., A.Y., L.G.), University of
| | - John G.F. Cleland
- From the Department of Cardiology (E.T.A., J.K., L.G.), Research Institute of Internal Medicine (E.T.A., S.N., T.U., A.Y., P.A.), Section of Clinical Immunology and Infectious Diseases (P.A.), Institute for Surgical Research (J.G.), Department of Medical Biochemistry, Oslo University Hospital Rikshospitalet (R.W.), Faculty of Medicine (T.U., J.K., A.Y., P.A., L.G.), and K.G. Jebsen Cardiac Research Centre and Center for Heart Failure Research (E.T.A., S.N., J.G., J.K., A.Y., L.G.), University of
| | - John J.V. McMurray
- From the Department of Cardiology (E.T.A., J.K., L.G.), Research Institute of Internal Medicine (E.T.A., S.N., T.U., A.Y., P.A.), Section of Clinical Immunology and Infectious Diseases (P.A.), Institute for Surgical Research (J.G.), Department of Medical Biochemistry, Oslo University Hospital Rikshospitalet (R.W.), Faculty of Medicine (T.U., J.K., A.Y., P.A., L.G.), and K.G. Jebsen Cardiac Research Centre and Center for Heart Failure Research (E.T.A., S.N., J.G., J.K., A.Y., L.G.), University of
| | - Pål Aukrust
- From the Department of Cardiology (E.T.A., J.K., L.G.), Research Institute of Internal Medicine (E.T.A., S.N., T.U., A.Y., P.A.), Section of Clinical Immunology and Infectious Diseases (P.A.), Institute for Surgical Research (J.G.), Department of Medical Biochemistry, Oslo University Hospital Rikshospitalet (R.W.), Faculty of Medicine (T.U., J.K., A.Y., P.A., L.G.), and K.G. Jebsen Cardiac Research Centre and Center for Heart Failure Research (E.T.A., S.N., J.G., J.K., A.Y., L.G.), University of
| | - Lars Gullestad
- From the Department of Cardiology (E.T.A., J.K., L.G.), Research Institute of Internal Medicine (E.T.A., S.N., T.U., A.Y., P.A.), Section of Clinical Immunology and Infectious Diseases (P.A.), Institute for Surgical Research (J.G.), Department of Medical Biochemistry, Oslo University Hospital Rikshospitalet (R.W.), Faculty of Medicine (T.U., J.K., A.Y., P.A., L.G.), and K.G. Jebsen Cardiac Research Centre and Center for Heart Failure Research (E.T.A., S.N., J.G., J.K., A.Y., L.G.), University of
| |
Collapse
|
181
|
Grothusen C, Schuett H, Hillmer A, Lumpe S, Grote K, Ballmaier M, Bleich A, Glage S, Tietge UJF, Luchtefeld M, Schieffer B. Role of suppressor of cytokine signaling-1 in murine atherosclerosis. PLoS One 2012; 7:e51608. [PMID: 23300554 PMCID: PMC3531439 DOI: 10.1371/journal.pone.0051608] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 11/02/2012] [Indexed: 01/26/2023] Open
Abstract
Background While the impact of inflammation as the substantial driving force of atherosclerosis has been investigated in detail throughout the years, the influence of negative regulators of pro-atherogenic pathways on plaque development has remained largely unknown. Suppressor of cytokine signaling (SOCS)-1 potently restricts transduction of various inflammatory signals and, thereby modulates T-cell development, macrophage activation and dendritic cell maturation. Its role in atherogenesis, however has not been elucidated so far. Methods and Results Loss of SOCS-1 in the low-density lipoprotein receptor deficient murine model of atherosclerosis resulted in a complex, systemic and ultimately lethal inflammation with increased generation of Ly-6Chi monocytes and activated macrophages. Even short-term exposure of these mice to high-cholesterol dieting caused enhanced atherosclerotic plaque development with accumulation of M1 macrophages, Ly-6C positive cells and neutrophils. Conclusion Our data not only imply that SOCS-1 is athero-protective but also emphasize the fundamental, regulatory importance of SOCS-1 in inflammation-prone organisms.
Collapse
Affiliation(s)
- Christina Grothusen
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Harald Schuett
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Anja Hillmer
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Stefan Lumpe
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Karsten Grote
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Matthias Ballmaier
- Department of Pediatric Haematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Andre Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Silke Glage
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Uwe J. F. Tietge
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Maren Luchtefeld
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Bernhard Schieffer
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
- * E-mail:
| |
Collapse
|
182
|
Plasmacytoid Dendritic Cells Play a Key Role in Promoting Atherosclerosis in Apolipoprotein E–Deficient Mice. Arterioscler Thromb Vasc Biol 2012; 32:2569-79. [DOI: 10.1161/atvbaha.112.251314] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Objective—
Clinical studies have identified that reduced numbers of circulating plasmacytoid dendritic cells (pDCs) act as a predictor of cardiovascular events in coronary artery disease and that pDCs are detectable in the shoulder region of human atherosclerotic plaques, where rupture is most likely to occur. Results from animal models are controversial, with pDCs seen to inhibit or promote lesion development depending on the experimental settings. Here, we investigated the role of pDCs in atherosclerosis in apolipoprotein E−deficient mice.
Methods and Results—
We demonstrated that the aorta and spleen of both apolipoprotein E−deficient and C57BL/6 mice displayed similar numbers of pDCs, with similar activation status. In contrast, assessment of antigen uptake/presentation using the Eα/Y-Ae system revealed that aortic pDCs in apolipoprotein E−deficient
-
mice were capable of presenting in vivo systemically administered antigen. Continuous treatment of apolipoprotein E−deficient mice with anti−mouse plasmacytoid dendritic cell antigen 1 (mPDCA-1) antibody caused specific depletion of pDCs in the aorta and spleen and significantly reduced atherosclerosis formation in the aortic sinus (by 46%;
P
<0.001). Depletion of pDCs also reduced macrophages (by 34%;
P
<0.05) and increased collagen content (by 41%;
P
<0.05) in aortic plaques, implying a more stable plaque phenotype. Additionally, pDC depletion reduced splenic T-cell activation and inhibited interleukin-12, chemokine (C-X-C motif) ligand 1, monokine induced by interferon-γ, interferon γ−induced protein 10, and vascular endothelium growth factor serum levels.
Conclusion—
These results identify a critical role for pDCs in atherosclerosis and suggest a potential role for pDC targeting in the control of the pathology.
Collapse
|
183
|
Rose-John S. IL-6 trans-signaling via the soluble IL-6 receptor: importance for the pro-inflammatory activities of IL-6. Int J Biol Sci 2012; 8:1237-47. [PMID: 23136552 PMCID: PMC3491447 DOI: 10.7150/ijbs.4989] [Citation(s) in RCA: 745] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 08/15/2012] [Indexed: 02/06/2023] Open
Abstract
Interleukin-6 (IL-6) is a cytokine with many activities. It has functions in the regulation of the immune system and the nervous system. Furthermore, IL-6 is involved in liver regeneration and in the metabolic control of the body. On target cells, IL-6 binds to an 80 kDa IL-6 receptor (IL-6R). The complex of IL-6 and IL-6R associates with a second protein, gp130, which thereupon dimerizes and initiates intracellular signaling. Whereas gp130 is expressed on all cells, IL-6R is only present on few cells in the body including hepatocytes and some leukocytes. Cells, which do not express IL-6R cannot respond to the cytokine, since gp130 alone has no measurable affinity for IL-6. Interestingly, a soluble form of IL-6R (sIL-6R) comprising the extracellular portion of the receptor can bind IL-6 with a similar affinity as the membrane bound IL-6R. The complex of IL-6 and sIL-6R can bind to gp130 on cells, which do not express the IL-6R, and which are unresponsive to IL-6. This process has been called trans-signaling. Here I will review published evidence that IL-6 trans-signaling is pro-inflammatory whereas classic IL-6 signaling via the membrane bound IL-6R is needed for regenerative or anti-inflammatory activities of the cytokine. Furthermore, the detailed knowledge of IL-6 biology has important consequences for therapeutic strategies aimed at the blockade of the cytokine IL-6.
Collapse
Affiliation(s)
- Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-University of Kiel, Kiel, Germany.
| |
Collapse
|
184
|
Kahlenberg JM, Kaplan MJ. Mechanisms of premature atherosclerosis in rheumatoid arthritis and lupus. Annu Rev Med 2012; 64:249-63. [PMID: 23020882 DOI: 10.1146/annurev-med-060911-090007] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE), the two most common systemic autoimmune disorders, have both unique and overlapping manifestations. One feature they share is a significantly enhanced risk of atherosclerotic cardiovascular (CV) disease that significantly contributes to morbidity and mortality. The primary mechanisms that drive CV damage in these diseases remain to be fully characterized, but recent discoveries indicate that distinct inflammatory pathways and immune dysregulation characteristic of RA and SLE likely play prominent roles. This review focuses on analyzing the major mechanisms and pathways potentially implicated in the acceleration of atherothrombosis and CV risk in SLE and RA, as well as in the identification of putative preventive strategies that may mitigate vascular complications in systemic autoimmunity.
Collapse
Affiliation(s)
- J Michelle Kahlenberg
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109-5680, USA.
| | | |
Collapse
|
185
|
|
186
|
Waetzig GH, Rose-John S. Hitting a complex target: an update on interleukin-6 trans-signalling. Expert Opin Ther Targets 2012; 16:225-36. [PMID: 22324903 DOI: 10.1517/14728222.2012.660307] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Interleukin-6 (IL-6) is a key target in inflammation and cancer. Selective inhibition of IL-6 trans-signalling could provide the same or even higher therapeutic efficacy with a better side effect profile than complete IL-6 inhibition. Animal studies with IL-6 inhibitors show that the classic IL-6 signalling pathway via the membrane-bound IL-6 receptor (IL-6R) has important physiological functions, whereas blocking the trans-signalling pathway via the soluble IL-6R (sIL-6R) is sufficient to prevent or treat IL-6-driven diseases. Due to the success of the anti-IL-6R antibody tocilizumab and difficulties of constructing selective trans-signalling inhibitors, most drug candidates in clinical development target IL-6 or IL-6R and, thus, both IL-6 pathways. By contrast, the fusion protein sgp130Fc selectively targets IL-6/sIL-6R trans-signalling by utilising the soluble gp130 receptor as the natural inhibitor of trans-signalling. AREAS COVERED The authors summarise recent developments in the field with a focus on animal studies highlighting the mechanistic differences between classic and trans-signalling and their therapeutic implications. EXPERT OPINION Characterising disease mechanisms in terms of the employed IL-6 pathways will help to select the right therapeutic IL-6 inhibitor in the future. The trans-signalling inhibitor sgp130Fc is about to enter the clinic and holds promise for a clinically different profile in comparison with complete IL-6 inhibitors.
Collapse
Affiliation(s)
- Georg H Waetzig
- Christian-Albrechts-University Kiel, Institute of Biochemistry, Kiel, Germany
| | | |
Collapse
|
187
|
Sarwar N, Butterworth AS, Freitag DF, Gregson J, Willeit P, Gorman DN, Gao P, Saleheen D, Rendon A, Nelson CP, Braund PS, Hall AS, Chasman DI, Tybjærg-Hansen A, Chambers JC, Benjamin EJ, Franks PW, Clarke R, Wilde AAM, Trip MD, Steri M, Witteman JCM, Qi L, van der Schoot CE, de Faire U, Erdmann J, Stringham HM, Koenig W, Rader DJ, Melzer D, Reich D, Psaty BM, Kleber ME, Panagiotakos DB, Willeit J, Wennberg P, Woodward M, Adamovic S, Rimm EB, Meade TW, Gillum RF, Shaffer JA, Hofman A, Onat A, Sundström J, Wassertheil-Smoller S, Mellström D, Gallacher J, Cushman M, Tracy RP, Kauhanen J, Karlsson M, Salonen JT, Wilhelmsen L, Amouyel P, Cantin B, Best LG, Ben-Shlomo Y, Manson JE, Davey-Smith G, de Bakker PIW, O'Donnell CJ, Wilson JF, Wilson AG, Assimes TL, Jansson JO, Ohlsson C, Tivesten Å, Ljunggren Ö, Reilly MP, Hamsten A, Ingelsson E, Cambien F, Hung J, Thomas GN, Boehnke M, Schunkert H, Asselbergs FW, Kastelein JJP, Gudnason V, Salomaa V, Harris TB, Kooner JS, Allin KH, Nordestgaard BG, Hopewell JC, Goodall AH, Ridker PM, Hólm H, Watkins H, Ouwehand WH, Samani NJ, Kaptoge S, Di Angelantonio E, Harari O, Danesh J. Interleukin-6 receptor pathways in coronary heart disease: a collaborative meta-analysis of 82 studies. Lancet 2012; 379:1205-13. [PMID: 22421339 PMCID: PMC3316940 DOI: 10.1016/s0140-6736(11)61931-4] [Citation(s) in RCA: 619] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Persistent inflammation has been proposed to contribute to various stages in the pathogenesis of cardiovascular disease. Interleukin-6 receptor (IL6R) signalling propagates downstream inflammation cascades. To assess whether this pathway is causally relevant to coronary heart disease, we studied a functional genetic variant known to affect IL6R signalling. METHODS In a collaborative meta-analysis, we studied Asp358Ala (rs2228145) in IL6R in relation to a panel of conventional risk factors and inflammation biomarkers in 125,222 participants. We also compared the frequency of Asp358Ala in 51,441 patients with coronary heart disease and in 136,226 controls. To gain insight into possible mechanisms, we assessed Asp358Ala in relation to localised gene expression and to postlipopolysaccharide stimulation of interleukin 6. FINDINGS The minor allele frequency of Asp358Ala was 39%. Asp358Ala was not associated with lipid concentrations, blood pressure, adiposity, dysglycaemia, or smoking (p value for association per minor allele ≥0·04 for each). By contrast, for every copy of 358Ala inherited, mean concentration of IL6R increased by 34·3% (95% CI 30·4-38·2) and of interleukin 6 by 14·6% (10·7-18·4), and mean concentration of C-reactive protein was reduced by 7·5% (5·9-9·1) and of fibrinogen by 1·0% (0·7-1·3). For every copy of 358Ala inherited, risk of coronary heart disease was reduced by 3·4% (1·8-5·0). Asp358Ala was not related to IL6R mRNA levels or interleukin-6 production in monocytes. INTERPRETATION Large-scale human genetic and biomarker data are consistent with a causal association between IL6R-related pathways and coronary heart disease. FUNDING British Heart Foundation; UK Medical Research Council; UK National Institute of Health Research, Cambridge Biomedical Research Centre; BUPA Foundation.
Collapse
|