1
|
Cederström S, Jernberg T, Samnegård A, Johansson F, Silveira A, Tornvall P, Lundman P. Inflammatory biomarkers and long-term outcome in young patients three months after a first myocardial infarction. Cytokine 2024; 182:156696. [PMID: 39059290 DOI: 10.1016/j.cyto.2024.156696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/18/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Studies on predictive value of circulating inflammatory biomarkers after myocardial infarction (MI) have often been limited by blood sampling only in an acute setting and short follow-up time. We aimed to compare the long-term predictive value of nine inflammatory biomarkers, known to be involved in atherosclerosis, in young patients investigated three months after a first-time MI. METHODS Nine biomarkers (high-sensitivity C-reactive protein, interleukin (IL)-6, IL-18, monocyte chemoattractant protein-1, matrix metalloproteinase (MMP)-1, MMP-3, MMP-9, serum amyloid A and tumor necrosis factor-alfa) were sampled in 382 young (<60 years) patients and in age and sex-matched controls, three months after a first-time MI between 1996 and 2000. Swedish national patient registers were used to determine cardiovascular (CV) outcomes during 20 years of follow-up. RESULTS In cases, random forest models identified IL-6 as the most important predictor of the primary composite endpoint of death, heart failure (HF) or MI hospitalization, and the separate endpoints death and HF hospitalization. IL-18 was the most important predictor of MI hospitalization. In a Cox regression, the highest tertile of IL-6 was associated with the composite endpoint (HR (95% CI) 1.91 (1.31-2.79)), death (2.38 (1.42-3.98)) and HF hospitalization (2.70 (1.32-5.50)), when adjusting for age, sex and CV risk factors. The highest tertile of IL-18 was associated with MI hospitalization (2.31 (1.08-4.91)) when severity of coronary atherosclerosis was added to the same type of model. CONCLUSIONS When nine inflammatory markers involved in atherosclerosis were analyzed three months after the acute event in young MI patients, IL-6 and IL-18 were the most important biomarkers to predict long-term CV outcomes during 20 years of follow-up.
Collapse
Affiliation(s)
- Sofia Cederström
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Sweden.
| | - Tomas Jernberg
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Sweden
| | - Ann Samnegård
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Sweden
| | - Fredrik Johansson
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Sweden
| | - Angela Silveira
- Department of Medicine Solna K2, Karolinska Institutet and Karolinska University Hospital Solna
| | - Per Tornvall
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Pia Lundman
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Sweden
| |
Collapse
|
2
|
González L, Rivera K, Andia ME, Martínez Rodriguez G. The IL-1 Family and Its Role in Atherosclerosis. Int J Mol Sci 2022; 24:17. [PMID: 36613465 PMCID: PMC9820551 DOI: 10.3390/ijms24010017] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
The IL-1 superfamily of cytokines is a central regulator of immunity and inflammation. The family is composed of 11 cytokines (with agonist, antagonist, and anti-inflammatory properties) and 10 receptors, all tightly regulated through decoy receptor, receptor antagonists, and signaling inhibitors. Inflammation not only is an important physiological response against infection and injury but also plays a central role in atherosclerosis development. Several clinical association studies along with experimental studies have implicated the IL-1 superfamily of cytokines and its receptors in the pathogenesis of cardiovascular disease. Here, we summarize the key features of the IL-1 family, its role in immunity and disease, and how it helps shape the development of atherosclerosis.
Collapse
Affiliation(s)
- Leticia González
- Centro de Imágenes Biomédicas—Departamento de Radiología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago 3580000, Chile
- Instituto Milenio de Ingeniería e Inteligencia Artificial Para la Salud, iHEALTH, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Katherine Rivera
- Centro de Imágenes Biomédicas—Departamento de Radiología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago 3580000, Chile
- Programa de Doctorado en Ciencias Médicas, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 3580000, Chile
| | - Marcelo E. Andia
- Centro de Imágenes Biomédicas—Departamento de Radiología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago 3580000, Chile
- Instituto Milenio de Ingeniería e Inteligencia Artificial Para la Salud, iHEALTH, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Gonzalo Martínez Rodriguez
- División de Enfermedades Cardiovasculares, Pontificia Universidad Católica de Chile, Santiago 3580000, Chile
| |
Collapse
|
3
|
Liu L, Shi Z, Ji X, Zhang W, Luan J, Zahr T, Qiang L. Adipokines, adiposity, and atherosclerosis. Cell Mol Life Sci 2022; 79:272. [PMID: 35503385 PMCID: PMC11073100 DOI: 10.1007/s00018-022-04286-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/11/2022] [Accepted: 04/03/2022] [Indexed: 12/12/2022]
Abstract
Characterized by a surplus of whole-body adiposity, obesity is strongly associated with the prognosis of atherosclerosis, a hallmark of coronary artery disease (CAD) and the major contributor to cardiovascular disease (CVD) mortality. Adipose tissue serves a primary role as a lipid-storage organ, secreting cytokines known as adipokines that affect whole-body metabolism, inflammation, and endocrine functions. Emerging evidence suggests that adipokines can play important roles in atherosclerosis development, progression, as well as regression. Here, we review the versatile functions of various adipokines in atherosclerosis and divide these respective functions into three major groups: protective, deteriorative, and undefined. The protective adipokines represented here are adiponectin, fibroblast growth factor 21 (FGF-21), C1q tumor necrosis factor-related protein 9 (CTRP9), and progranulin, while the deteriorative adipokines listed include leptin, chemerin, resistin, Interleukin- 6 (IL-6), and more, with additional adipokines that have unclear roles denoted as undefined adipokines. Comprehensively categorizing adipokines in the context of atherosclerosis can help elucidate the various pathways involved and potentially pave novel therapeutic approaches to treat CVDs.
Collapse
Affiliation(s)
- Longhua Liu
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China.
| | - Zunhan Shi
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Xiaohui Ji
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Wenqian Zhang
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Jinwen Luan
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Tarik Zahr
- Department of Pharmacology, Columbia University, New York, NY, USA
| | - Li Qiang
- Department of Pathology and Cellular Biology and Naomi Berrie Diabetes Center, Columbia University, New York, NY, USA.
| |
Collapse
|
4
|
Rezaieyazdi Z, AkbariRad M, Saadati N, Salari M, Orang R, Sedighi S, Esmaily H, Azarpazhooh MR, Firoozi A, Akbarpour E. Serum interleukin-18 and its relationship with subclinical atherosclerosis in systemic lupus erythematosus. ARYA ATHEROSCLEROSIS 2021; 17:1-6. [PMID: 35685451 PMCID: PMC9145840 DOI: 10.22122/arya.v17i0.2126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 06/30/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND Interleukin-18 (IL-18) is a pro-inflammatory and pro-atherogenic factor, and its blood level has shown a direct correlation with atherosclerosis. We aimed to evaluate the serum IL-18 level in patients with systemic lupus erythematosus (SLE) and its relationship with the intima-media thickness (IMT) of the carotid artery in these patients, as an indicator of atherosclerosis. METHODS In this cross-sectional study, 60 patients as the patient group and 30 healthy volunteers as the control group [matched sex, age, and body mass index (BMI)] were selected, and their disease status and general data were gathered using the Systemic Lupus Erythematosus Disease Activity Index 2000 (SLEDAI-2K) form. A blood sample was also obtained from all participants to determine the serum level of IL-18 and other metrics, including high-sensitivity C-reactive protein (hs-CRP), cholesterol, triglyceride (TG), low-density lipoprotein (LDL), high-density lipoprotein (HDL), anti-double stranded deoxyribonucleic acid (anti-dsDNA), complement 3 (C3), and C4. The IMT of the carotid artery was calculated in both groups. We also evaluated the clinical cardiovascular manifestations. RESULTS The serum IL-18 levels in patients were significantly higher than in the control group (P ˂ 0.005). It had no significant correlation with disease activity (P = 0.10). The patients with SLE with high IL-18 serum levels (> 280 pg/ml) had higher SLEDAI-2K (P = 0.02) than the patients with a low level (< 280), where 280 was the median of the IL-18 levels. The serum IL-18 level had no significant correlation with the carotid artery IMT. CONCLUSION A high level of IL-18 reflects the disease activity, but it was not significantly correlated with subclinical atherosclerosis, denoted by the carotid artery IMT.
Collapse
Affiliation(s)
- Zahra Rezaieyazdi
- Rheumatic Diseases Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mina AkbariRad
- Assistant Professor, Department of Internal Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nayyereh Saadati
- Rheumatic Diseases Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Masoumeh Salari
- Rheumatic Diseases Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Orang
- Rheumatic Diseases Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sima Sedighi
- Golestan Rheumatology Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Habibollah Esmaily
- Department of Epidemiology and Biostatistics, School of Health, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Abdollah Firoozi
- Pharmacist, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ensieh Akbarpour
- Department of Epidemiology and Biostatistics, School of Health, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
5
|
Multiplex Protein Biomarker Profiling in Patients with Familial Hypercholesterolemia. Genes (Basel) 2021; 12:genes12101599. [PMID: 34680994 PMCID: PMC8535274 DOI: 10.3390/genes12101599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/29/2021] [Accepted: 10/08/2021] [Indexed: 11/17/2022] Open
Abstract
Familial hypercholesterolemia (FH), is an autosomal dominant disorder caused by mutations in the LDLR, APOB, PCSK9, and APOE genes and is characterized by high plasma levels of total and low-density lipoprotein (LDL) cholesterol. Our study aimed to analyze the influences of two different therapies on a wide spectrum of plasma protein biomarkers of cardiovascular diseases. Plasma from FH patients under hypolipidemic therapy (N = 18; men = 8, age 55.4 ± 13.1 years) and patients under combined long-term LDL apheresis/hypolipidemic therapy (N = 14; men = 7; age 58.0 ± 13.6 years) were analyzed in our study. We measured a profile of 184 cardiovascular disease (CVD) associated proteins using a proximity extension assay (PEA). Hypolipidemic therapy significantly (all p < 0.01) influenced 10 plasma proteins (TM, DKK1, CCL3, CD4, PDGF subunit B, AGRP, IL18, THPO, and LOX1 decreased; ST2 increased). Under combined apheresis/hypolipidemic treatment, 18 plasma proteins (LDLR, PCSK9, MMP-3, GDF2, CTRC, SORT1, VEGFD, IL27, CCL24, and KIM1 decreased; OPN, COL1A1, KLK6, IL4RA, PLC, TNFR1, GLO1, and PTX3 increased) were significantly affected (all p < 0.006). Hypolipidemic treatment mainly affected biomarkers involved in vascular endothelial maintenance. Combined therapy influenced proteins that participate in cholesterol metabolism and inflammation.
Collapse
|
6
|
Ridker PM, MacFadyen JG, Thuren T, Libby P. Residual inflammatory risk associated with interleukin-18 and interleukin-6 after successful interleukin-1β inhibition with canakinumab: further rationale for the development of targeted anti-cytokine therapies for the treatment of atherothrombosis. Eur Heart J 2021; 41:2153-2163. [PMID: 31504417 DOI: 10.1093/eurheartj/ehz542] [Citation(s) in RCA: 141] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/05/2019] [Accepted: 07/26/2019] [Indexed: 11/15/2022] Open
Abstract
AIMS The Canakinumab Antiinflammatory Thrombosis Outcomes Study (CANTOS) established that targeting inflammation with interleukin-1β (IL-1β) inhibition can significantly reduce cardiovascular (CV) event rates in the absence of any beneficial effects on cholesterol. Yet, CANTOS participants treated with both high-intensity statins and canakinumab remain at considerable risk for recurrent CV events. Both interleukin-18 (IL-18, which like IL-1β requires the NLRP3 inflammasome for activation) and interleukin-6 (IL-6, a pro-inflammatory cytokine downstream of IL-1) may contribute to the recurrent events that occur even on canakinumab therapy, and thus represent novel targets for treating atherothrombosis. METHODS AND RESULTS Plasma samples from 4848 stable post-myocardial infarction patients who were assigned to active IL-1β inhibition or placebo within CANTOS underwent measurement of IL-18 and IL-6 both before and after initiation of canakinumab using validated ELISA. All participants were followed over a median 3.7-year period (maximum 5 years) for recurrent major adverse cardiovascular events (MACE) and for all-cause mortality. Compared to placebo, canakinumab significantly reduced IL-6 levels in a dose-dependent manner yielding placebo-subtracted median percent reductions in IL-6 at 3 months of 24.8%, 36.3%, and 43.2% for the 50, 150, and 300 mg doses, respectively (all P-values <0.001). By contrast, no dose of canakinumab significantly altered IL-18 levels measured at 3 months (all effects <1%, all P-values > 0.05). Yet, despite these differential plasma effects, either baseline and on-treatment levels of IL-18 or IL-6 associated with rates of future CV events. For example, for MACE, each tertile increase in IL-18 measured 3 months after canakinumab initiation associated with a 15% increase in risk [95% confidence interval (CI) 3-29%, P = 0.016], while each tertile increase in IL-6 measured 3 months after canakinumab initiation associated with a 42% increase in risk (95% CI 26-59%, P < 0.0001). Similar effects were observed for MACE-plus, CV death, all-cause mortality, and the for the combination endpoint of all vascular events inclusive of revascularization procedures and hospitalization for congestive heart failure. In baseline as well as on-treatment analyses, risks were highest among those with the highest levels of both IL-18 and IL-6. CONCLUSION There remains substantial residual inflammatory risk related to both IL-18 and IL-6 after IL-1β inhibition with canakinumab These data support further pharmacologic development of therapies for atherothrombosis that target IL-18 or IL-6 signalling, or that can simultaneously inhibit both IL-1β and IL-18 (such as NLRP3 inflammasome inhibitors). CLINICAL TRIAL REGISTRATION ClinicalTrials.gov NCT01327846.
Collapse
Affiliation(s)
- Paul M Ridker
- Department of Medicine, Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Harvard Medical School, 900 Commonwealth Avenue, Boston, MA 02215, USA.,Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Jean G MacFadyen
- Department of Medicine, Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Harvard Medical School, 900 Commonwealth Avenue, Boston, MA 02215, USA
| | - Tom Thuren
- Novartis Pharmaceutical Corporation, One Health Plaza, East Hanover, NJ 07936, USA
| | - Peter Libby
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| |
Collapse
|
7
|
Medina-Leyte DJ, Zepeda-García O, Domínguez-Pérez M, González-Garrido A, Villarreal-Molina T, Jacobo-Albavera L. Endothelial Dysfunction, Inflammation and Coronary Artery Disease: Potential Biomarkers and Promising Therapeutical Approaches. Int J Mol Sci 2021; 22:3850. [PMID: 33917744 PMCID: PMC8068178 DOI: 10.3390/ijms22083850] [Citation(s) in RCA: 193] [Impact Index Per Article: 64.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 12/14/2022] Open
Abstract
Coronary artery disease (CAD) and its complications are the leading cause of death worldwide. Inflammatory activation and dysfunction of the endothelium are key events in the development and pathophysiology of atherosclerosis and are associated with an elevated risk of cardiovascular events. There is great interest to further understand the pathophysiologic mechanisms underlying endothelial dysfunction and atherosclerosis progression, and to identify novel biomarkers and therapeutic strategies to prevent endothelial dysfunction, atherosclerosis and to reduce the risk of developing CAD and its complications. The use of liquid biopsies and new molecular biology techniques have allowed the identification of a growing list of molecular and cellular markers of endothelial dysfunction, which have provided insight on the molecular basis of atherosclerosis and are potential biomarkers and therapeutic targets for the prevention and or treatment of atherosclerosis and CAD. This review describes recent information on normal vascular endothelium function, as well as traditional and novel potential biomarkers of endothelial dysfunction and inflammation, and pharmacological and non-pharmacological therapeutic strategies aimed to protect the endothelium or reverse endothelial damage, as a preventive treatment for CAD and related complications.
Collapse
Affiliation(s)
- Diana Jhoseline Medina-Leyte
- Genomics of Cardiovascular Diseases Laboratory, National Institute of Genomic Medicine (INMEGEN), Mexico City 14610, Mexico; (D.J.M.-L.); (O.Z.-G.); (M.D.-P.); (A.G.-G.); (T.V.-M.)
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México (UNAM), Coyoacán, Mexico City 04510, Mexico
| | - Oscar Zepeda-García
- Genomics of Cardiovascular Diseases Laboratory, National Institute of Genomic Medicine (INMEGEN), Mexico City 14610, Mexico; (D.J.M.-L.); (O.Z.-G.); (M.D.-P.); (A.G.-G.); (T.V.-M.)
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México (UNAM), Coyoacán, Mexico City 04510, Mexico
| | - Mayra Domínguez-Pérez
- Genomics of Cardiovascular Diseases Laboratory, National Institute of Genomic Medicine (INMEGEN), Mexico City 14610, Mexico; (D.J.M.-L.); (O.Z.-G.); (M.D.-P.); (A.G.-G.); (T.V.-M.)
| | - Antonia González-Garrido
- Genomics of Cardiovascular Diseases Laboratory, National Institute of Genomic Medicine (INMEGEN), Mexico City 14610, Mexico; (D.J.M.-L.); (O.Z.-G.); (M.D.-P.); (A.G.-G.); (T.V.-M.)
| | - Teresa Villarreal-Molina
- Genomics of Cardiovascular Diseases Laboratory, National Institute of Genomic Medicine (INMEGEN), Mexico City 14610, Mexico; (D.J.M.-L.); (O.Z.-G.); (M.D.-P.); (A.G.-G.); (T.V.-M.)
| | - Leonor Jacobo-Albavera
- Genomics of Cardiovascular Diseases Laboratory, National Institute of Genomic Medicine (INMEGEN), Mexico City 14610, Mexico; (D.J.M.-L.); (O.Z.-G.); (M.D.-P.); (A.G.-G.); (T.V.-M.)
| |
Collapse
|
8
|
A novel anti-human IL-1R7 antibody reduces IL-18-mediated inflammatory signaling. J Biol Chem 2021; 296:100630. [PMID: 33823154 PMCID: PMC8018910 DOI: 10.1016/j.jbc.2021.100630] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/29/2021] [Accepted: 04/01/2021] [Indexed: 12/17/2022] Open
Abstract
Unchecked inflammation can result in severe diseases with high mortality, such as macrophage activation syndrome (MAS). MAS and associated cytokine storms have been observed in COVID-19 patients exhibiting systemic hyperinflammation. Interleukin-18 (IL-18), a proinflammatory cytokine belonging to the IL-1 family, is elevated in both MAS and COVID-19 patients, and its level is known to correlate with the severity of COVID-19 symptoms. IL-18 binds its specific receptor IL-1 receptor 5 (IL-1R5, also known as IL-18 receptor alpha chain), leading to the recruitment of the coreceptor, IL-1 receptor 7 (IL-1R7, also known as IL-18 receptor beta chain). This heterotrimeric complex then initiates downstream signaling, resulting in systemic and local inflammation. Here, we developed a novel humanized monoclonal anti-IL-1R7 antibody to specifically block the activity of IL-18 and its inflammatory signaling. We characterized the function of this antibody in human cell lines, in freshly obtained peripheral blood mononuclear cells (PBMCs) and in human whole blood cultures. We found that the anti-IL-1R7 antibody significantly suppressed IL-18-mediated NFκB activation, reduced IL-18-stimulated IFNγ and IL-6 production in human cell lines, and reduced IL-18-induced IFNγ, IL-6, and TNFα production in PBMCs. Moreover, the anti-IL-1R7 antibody significantly inhibited LPS- and Candida albicans–induced IFNγ production in PBMCs, as well as LPS-induced IFNγ production in whole blood cultures. Our data suggest that blocking IL-1R7 could represent a potential therapeutic strategy to specifically modulate IL-18 signaling and may warrant further investigation into its clinical potential for treating IL-18-mediated diseases, including MAS and COVID-19.
Collapse
|
9
|
Wienke J, Mertens JS, Garcia S, Lim J, Wijngaarde CA, Yeo JG, Meyer A, van den Hoogen LL, Tekstra J, Hoogendijk JE, Otten HG, Fritsch-Stork RDE, de Jager W, Seyger MMB, Thurlings RM, de Jong EMGJ, van der Kooi AJ, van der Pol WL, Arkachaisri T, Radstake TRDJ, van Royen-Kerkhof A, van Wijk F. Biomarker profiles of endothelial activation and dysfunction in rare systemic autoimmune diseases: implications for cardiovascular risk. Rheumatology (Oxford) 2021; 60:785-801. [PMID: 32810267 DOI: 10.1093/rheumatology/keaa270] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 03/19/2020] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVES Vasculopathy is an important hallmark of systemic chronic inflammatory connective tissue diseases (CICTD) and is associated with increased cardiovascular risk. We investigated disease-specific biomarker profiles associated with endothelial dysfunction, angiogenic homeostasis and (tissue) inflammation, and their relation to disease activity in rare CICTD. METHODS A total of 38 serum proteins associated with endothelial (dys)function and inflammation were measured by multiplex-immunoassay in treatment-naive patients with localized scleroderma (LoS, 30), eosinophilic fasciitis (EF, 8) or (juvenile) dermatomyositis (34), 119 (follow-up) samples during treatment, and 65 controls. Data were analysed by unsupervised clustering, Spearman correlations, non-parametric t test and ANOVA. RESULTS The systemic CICTD, EF and dermatomyositis, had distinct biomarker profiles, with 'signature' markers galectin-9 (dermatomyositis) and CCL4, CCL18, CXCL9, fetuin, fibronectin, galectin-1 and TSP-1 (EF). In LoS, CCL18, CXCL9 and CXCL10 were subtly increased. Furthermore, dermatomyositis and EF shared upregulation of markers related to interferon (CCL2, CXCL10), endothelial activation (VCAM-1), inhibition of angiogenesis (angiopoietin-2, sVEGFR-1) and inflammation/leucocyte chemo-attraction (CCL19, CXCL13, IL-18, YKL-40), as well as disturbance of the Angiopoietin-Tie receptor system and VEGF-VEGFR system. These profiles were related to disease activity, and largely normalized during treatment. However, a subgroup of CICTD patients showed continued elevation of CXCL10, CXCL13, galectin-9, IL-18, TNFR2, VCAM-1, and/or YKL-40 during clinically inactive disease, possibly indicating subclinical interferon-driven inflammation and/or endothelial dysfunction. CONCLUSION CICTD-specific biomarker profiles revealed an anti-angiogenic, interferon-driven environment during active disease, with incomplete normalization under treatment. This warrants further investigation into monitoring of vascular biomarkers during clinical follow-up, or targeted interventions to minimize cardiovascular risk in the long term.
Collapse
Affiliation(s)
- Judith Wienke
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jorre S Mertens
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands.,Department of Dermatology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Samuel Garcia
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Johan Lim
- Department of Neurology, Amsterdam University Medical Centre, University of Amsterdam, Neuroscience Institute, Amsterdam, Netherlands
| | - Camiel A Wijngaarde
- Department of Neurology and Neurosurgery, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Joo Guan Yeo
- Rheumatology and Immunology Service, Department of Paediatric Subspecialties, KK Women's and Children's Hospital and Duke-NUS Medical School, Duke, NUS, Singapore.,Translational Immunology Institute, SingHealth-Academic Medical Centre, Duke, NUS, Singapore
| | - Alain Meyer
- Service de Physiologie et d'Explorations Fonctionnelles, Centre, de Référence des, Maladies Autoimmunes Rares, Rhumatologie, Institut de Physiologie, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, France
| | - Lucas L van den Hoogen
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Janneke Tekstra
- Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Jessica E Hoogendijk
- Department of Neurology and Neurosurgery, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Henny G Otten
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Ruth D E Fritsch-Stork
- Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands.,Sigmund Freud Private University, Vienna, Austria, Vienna, Austria.,Medizinische Abteilung Hanusch Krankenhaus und Ludwig Boltzmann Institut für Osteologie, Vienna, Austria
| | - Wilco de Jager
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marieke M B Seyger
- Department of Dermatology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Rogier M Thurlings
- Department of Rheumatic Diseases, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Elke M G J de Jong
- Department of Dermatology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Anneke J van der Kooi
- Department of Neurology, Amsterdam University Medical Centre, University of Amsterdam, Neuroscience Institute, Amsterdam, Netherlands
| | - W Ludo van der Pol
- Department of Neurology and Neurosurgery, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | - Thaschawee Arkachaisri
- Rheumatology and Immunology Service, Department of Paediatric Subspecialties, KK Women's and Children's Hospital and Duke-NUS Medical School, Duke, NUS, Singapore
| | - Timothy R D J Radstake
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Annet van Royen-Kerkhof
- Paediatric Rheumatology and Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Femke van Wijk
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
10
|
Takahashi M. NLRP3 inflammasome as a key driver of vascular disease. Cardiovasc Res 2021; 118:372-385. [PMID: 33483732 DOI: 10.1093/cvr/cvab010] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/12/2020] [Accepted: 01/16/2021] [Indexed: 12/12/2022] Open
Abstract
NLRP3 (nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3) is an intracellular innate immune receptor that recognizes a diverse range of stimuli derived from pathogens, damaged or dead cells, and irritants. NLRP3 activation causes the assembly of a large multiprotein complex termed the NLRP3 inflammasome, and leads to the secretion of bioactive interleukin (IL)-1β and IL-18 as well as the induction of inflammatory cell death termed pyroptosis. Accumulating evidence indicates that NLRP3 inflammasome plays a key role in the pathogenesis of sterile inflammatory diseases, including atherosclerosis and other vascular diseases. Indeed, the results of the Canakinumab Anti-inflammatory Thrombosis Outcome Study (CANTOS) trial demonstrated that IL-1β-mediated inflammation plays an important role in atherothrombotic events and suggested that NLRP3 inflammasome is a key driver of atherosclerosis. In this review, we will summarize the current state of knowledge regarding the role of NLRP3 inflammasome in vascular diseases, in particular in atherosclerosis, vascular injury, aortic aneurysm, and Kawasaki disease vasculitis, and discuss NLRP3 inflammasome as a therapeutic target for these disorders.
Collapse
Affiliation(s)
- Masafumi Takahashi
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
11
|
Association between polymorphisms in interleukin-18 promoter and risk of coronary artery disease: a meta-analysis. Biosci Rep 2020; 39:220955. [PMID: 31661113 PMCID: PMC6863765 DOI: 10.1042/bsr20192721] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/17/2019] [Accepted: 10/25/2019] [Indexed: 12/11/2022] Open
Abstract
Background: Previous studies have explored associations between interleukin-18 (IL-18) promoter polymorphisms and coronary artery disease (CAD). However, the results were controversial. We conducted a meta-analysis to clarify the association between the two polymorphisms and CAD risk. Methods: We searched English and Chinese databases and calculated the odds ratio (OR) and 95% confidence interval (CI) to estimate whether there are genetic associations between IL-18 promoter polymorphisms and the risk of CAD. All relevant studies were screened and meta-analyzed using STATA 15.0. Results: A total of 15 studies, including 12 studies for -137 G/C and 9 studies for -607 C/A, were identified for the meta-analysis. For -137 G/C, the results showed a significantly reduced risk of CAD in the dominant model (OR = 0.85) and heterozygous model (OR = 0.88) in the overall analysis. However, in subgroup analysis, decreased CAD risks were only observed in Asian populations for heterozygous genetic models. For -607 C/A, the overall OR revealed a reduced risk of CAD in all five genetic models (allelic, OR = 0.78; recessive, OR = 0.75; dominant, OR = 0.68; homozygous, OR = 0.61; heterozygous, OR = 0.72). In subgroup analysis, reduced CAD risk was also found in five genetic models of the Asian population. We also found that the IL-18 polymorphisms were correlated with myocardial infarction (MI) and multivessel (MV) disease. Conclusion: Our results suggested that the -137 polymorphism and -607 polymorphism in the IL-18 promoter were negatively associated with CAD, especially in the Asian population. In addition, some genetic models were correlated with the severity of CAD.
Collapse
|
12
|
Oliveira MS, Rheinheimer J, Moehlecke M, Rodrigues M, Assmann TS, Leitão CB, Trindade MRM, Crispim D, de Souza BM. UCP2, IL18, and miR-133a-3p are dysregulated in subcutaneous adipose tissue of patients with obesity. Mol Cell Endocrinol 2020; 509:110805. [PMID: 32251712 DOI: 10.1016/j.mce.2020.110805] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 03/26/2020] [Accepted: 03/30/2020] [Indexed: 01/12/2023]
Abstract
The aim of this study was to compare the expression of UCP2, NLRP3, IL1B, IL18, and miR-133a-3p in subcutaneous adipose tissue (SAT) of 61 patients divided according to BMI: Group 1 (n = 8; BMI<25.0 kg/m2), Group 2 (n = 24; BMI 30.0-39.9 kg/m2), and Group 3 (n = 29; BMI≥40.0 kg/m2). SAT biopsies were obtained from individuals who underwent bariatric surgery or elective abdominal surgery. Gene expressions were quantified using qPCR. Bioinformatics analyses were employed to investigate target genes and pathways related to miR-133a-3p. UCP2 and miR-133a-3p expressions were decreased in SAT of Groups 2 and 3 while IL18 was increased compared to Group 1. NLRP3 and IL1B expressions did not differ between groups; however, NLRP3 was positively correlated with waist circumference and excess weight. Bioinformatics analysis demonstrated that UCP2 and NLRP3 are targets of miR-133a-3p. In conclusion, UCP2 and miR-133a-3p expressions are downregulated in patients with obesity, while IL18 is upregulated. NRLP3 is correlated with waist circumference and weight excess.
Collapse
Affiliation(s)
- Mayara S Oliveira
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Graduate Program of Medical Sciences: Endocrinology, Brazil
| | - Jakeline Rheinheimer
- Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Graduate Program of Medical Sciences: Endocrinology, Brazil
| | - Milene Moehlecke
- Department of Endocrinology, Universidade Luterana do Brasil, Canoas, Rio Grande do Sul, Brazil
| | - Michelle Rodrigues
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Taís S Assmann
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Graduate Program of Medical Sciences: Endocrinology, Brazil
| | - Cristiane B Leitão
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Graduate Program of Medical Sciences: Endocrinology, Brazil
| | - Manoel R M Trindade
- Digestive Surgery Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Daisy Crispim
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Graduate Program of Medical Sciences: Endocrinology, Brazil
| | - Bianca M de Souza
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Graduate Program of Medical Sciences: Endocrinology, Brazil.
| |
Collapse
|
13
|
Bartlett B, Ludewick HP, Misra A, Lee S, Dwivedi G. Macrophages and T cells in atherosclerosis: a translational perspective. Am J Physiol Heart Circ Physiol 2019; 317:H375-H386. [DOI: 10.1152/ajpheart.00206.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Atherosclerosis is now considered a chronic maladaptive inflammatory disease. The hallmark feature in both human and murine disease is atherosclerotic plaques. Macrophages and various T-cell lineages play a crucial role in atherosclerotic plaque establishment and disease progression. Humans and mice share many of the same processes that occur within atherogenesis. The various similarities enable considerable insight into disease mechanisms and those which contribute to cardiovascular complications. The apolipoprotein E-null and low-density lipoprotein receptor-null mice have served as the foundation for further immunological pathway manipulation to identify pro- and antiatherogenic pathways in attempt to reveal more novel therapeutic targets. In this review, we provide a translational perspective and discuss the roles of macrophages and various T-cell lineages in contrasting proatherosclerotic and atheroprotective settings.
Collapse
Affiliation(s)
- Benjamin Bartlett
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Murdoch, Western Australia, Australia
- School of Medicine, University of Western Australia, Perth, Western Australia, Australia
| | - Herbert P. Ludewick
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Murdoch, Western Australia, Australia
| | - Ashish Misra
- Heart Research Institute, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Silvia Lee
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Murdoch, Western Australia, Australia
- Department of Microbiology, Pathwest Laboratory Medicine, Perth, Western Australia, Australia
| | - Girish Dwivedi
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Murdoch, Western Australia, Australia
- School of Medicine, University of Western Australia, Perth, Western Australia, Australia
- Department of Cardiology, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| |
Collapse
|
14
|
Interleukin-18 in Health and Disease. Int J Mol Sci 2019; 20:ijms20030649. [PMID: 30717382 PMCID: PMC6387150 DOI: 10.3390/ijms20030649] [Citation(s) in RCA: 322] [Impact Index Per Article: 64.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 12/12/2022] Open
Abstract
Interleukin (IL)-18 was originally discovered as a factor that enhanced IFN-γ production from anti-CD3-stimulated Th1 cells, especially in the presence of IL-12. Upon stimulation with Ag plus IL-12, naïve T cells develop into IL-18 receptor (IL-18R) expressing Th1 cells, which increase IFN-γ production in response to IL-18 stimulation. Therefore, IL-12 is a commitment factor that induces the development of Th1 cells. In contrast, IL-18 is a proinflammatory cytokine that facilitates type 1 responses. However, IL-18 without IL-12 but with IL-2, stimulates NK cells, CD4+ NKT cells, and established Th1 cells, to produce IL-3, IL-9, and IL-13. Furthermore, together with IL-3, IL-18 stimulates mast cells and basophils to produce IL-4, IL-13, and chemical mediators such as histamine. Therefore, IL-18 is a cytokine that stimulates various cell types and has pleiotropic functions. IL-18 is a member of the IL-1 family of cytokines. IL-18 demonstrates a unique function by binding to a specific receptor expressed on various types of cells. In this review article, we will focus on the unique features of IL-18 in health and disease in experimental animals and humans.
Collapse
|
15
|
Abstract
Initially described as an interferon (IFN)γ‐inducing factor, interleukin (IL)‐18 is indeed involved in Th1 and NK cell activation, but also in Th2, IL‐17‐producing γδ T cells and macrophage activation. IL‐18, a member of the IL‐1 family, is similar to IL‐1β for being processed by caspase 1 to an 18 kDa‐biologically active mature form. IL‐18 binds to its specific receptor (IL‐18Rα, also known as IL‐1R7) forming a low affinity ligand chain. This is followed by recruitment of the IL‐18Rβ chain. IL‐18 then uses the same signaling pathway as IL‐1 to activate NF‐kB and induce inflammatory mediators such as adhesion molecules, chemokines and Fas ligand. IL‐18 also binds to the circulating high affinity IL‐18 binding protein (BP), such as only unbound free IL‐18 is active. IL‐18Rα may also bind IL‐37, another member of the IL‐1 family, but in association with the negative signaling chain termed IL‐1R8, which transduces an anti‐inflammatory signal. IL‐18BP also binds IL‐37 and this acts as a sink for the anti‐inflammatory properties of IL‐37. There is now ample evidence for a role of IL‐18 in various infectious, metabolic or inflammatory diseases such as influenza virus infection, atheroma, myocardial infarction, chronic obstructive pulmonary disease, or Crohn's disease. However, IL‐18 plays a very specific role in the pathogenesis of hemophagocytic syndromes (HS) also termed Macrophage Activation Syndrome. In children affected by NLRC4 gain‐of‐function mutations, IL‐18 circulates in the range of tens of nanograms/mL. HS is treated with the IL‐1 Receptor antagonist (anakinra) but also specifically with IL‐18BP. Systemic juvenile idiopathic arthritis or adult‐onset Still's disease are also characterized by high serum IL‐18 concentrations and are treated by IL‐18BP.
Collapse
Affiliation(s)
- Gilles Kaplanski
- Assistance Publique-Hôpitaux de Marseille, Centre Hospitalier Universitaire Conception, Service de Médecine Interne et Immunologie Clinique, Aix-Marseille Université, Marseille, France.,Vascular Research Center Marseille, Faculté de Pharmacie, Aix-Marseille Université, INSERM UMR_S1076, Marseille, France
| |
Collapse
|
16
|
ÖZzbïçer S, Uluçam ZM. Association Between Interleukin-18 Level and Left Ventricular Mass Index in Hypertensive Patients. Korean Circ J 2017; 47:238-244. [PMID: 28382080 PMCID: PMC5378031 DOI: 10.4070/kcj.2016.0351] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 10/24/2016] [Accepted: 11/01/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND OBJECTIVES In clinical trials, hypertensive patients tend to have higher interleukin-18 (IL-18) concentrations than normotensive groups, but the relationship between IL-18 and left ventricular hypertrophy (LVH), which is a marker of end-organ damage, is not well studied. We aimed to investigate the relationship between IL-18 and LVH in apparently healthy subjects free of clinically significant atherosclerotic disease. SUBJECTS AND METHODS We enrolled 198 subjects (102 women and 96 men) between May 2006 and March 2007, who were free of cardiovascular or immune diseases, but were suspected to have hypertension. Twenty-four-hour ambulatory blood pressure monitoring and two-dimensional echocardiography were performed. Lipid profiles, high-sensitivity CRP (hs-CRP), IL-18, and whole blood cell counts were measured for all subjects. RESULTS White blood cell count, hs-CRP, left ventricular mass, left ventricular mass index (LVMI), and IL-18 were higher in the hypertensive group than in the normotensive group (p=0.045, p=0.004, p<0.0001, p=0.001, and p=0.017 respectively). Twenty-four hour day and night systolic and diastolic blood pressure averages were positively correlated with IL-18 level in the entire study population. In multivariate regression analysis, left ventricular mass index and hs-CRP level were independently associated with IL-18 level in both the hypertensive group and the entire study population (β=0.154, β=0.149 p=0.033, p=0.040 and β=0.151, β=0.155 p=0.036, p=0.032 respectively). CONCLUSION We found that IL-18 level independently predicted LVMI in both the general population and in newly diagnosed hypertensive patients.
Collapse
Affiliation(s)
- Süleyman ÖZzbïçer
- Department of Cardiology, Adana Numune Training and Research Hospital, Adana, Turkey
| | | |
Collapse
|
17
|
Paramel Varghese G, Folkersen L, Strawbridge RJ, Halvorsen B, Yndestad A, Ranheim T, Krohg-Sørensen K, Skjelland M, Espevik T, Aukrust P, Lengquist M, Hedin U, Jansson JH, Fransén K, Hansson GK, Eriksson P, Sirsjö A. NLRP3 Inflammasome Expression and Activation in Human Atherosclerosis. J Am Heart Assoc 2016; 5:JAHA.115.003031. [PMID: 27207962 PMCID: PMC4889178 DOI: 10.1161/jaha.115.003031] [Citation(s) in RCA: 210] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background The NLR family, pyrin domain containing 3 (NLRP3) inflammasome is an interleukin (IL)‐1β and IL‐18 cytokine processing complex that is activated in inflammatory conditions. The role of the NLRP3 inflammasome in the pathogenesis of atherosclerosis and myocardial infarction is not fully understood. Methods and Results Atherosclerotic plaques were analyzed for transcripts of the NLRP3 inflammasome, and for IL‐1β release. The Swedish First‐ever myocardial Infarction study in Ac‐county (FIA) cohort consisting of DNA from 555 myocardial infarction patients and 1016 healthy individuals was used to determine the frequency of 4 single nucleotide polymorphisms (SNPs) from the downstream regulatory region of NLRP3. Expression of NLRP3, Apoptosis‐associated speck‐like protein containing a CARD (ASC), caspase‐1 (CASP1), IL1B, and IL18 mRNA was significantly increased in atherosclerotic plaques compared to normal arteries. The expression of NLRP3 mRNA was significantly higher in plaques of symptomatic patients when compared to asymptomatic ones. CD68‐positive macrophages were observed in the same areas of atherosclerotic lesions as NLRP3 and ASC expression. Occasionally, expression of NLRP3 and ASC was also present in smooth muscle cells. Cholesterol crystals and ATP induced IL‐1β release from lipopolysaccharide‐primed human atherosclerotic lesion plaques. The minor alleles of the variants rs4266924, rs6672995, and rs10733113 were associated with NLRP3 mRNA levels in peripheral blood mononuclear cells but not with the risk of myocardial infarction. Conclusions Our results indicate a possible role of the NLRP3 inflammasome and its genetic variants in the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Geena Paramel Varghese
- Cardiovascular Research Centre, Faculty of Medicine and Health, School of Health and Medical Sciences, Örebro University, Örebro, Sweden
| | - Lasse Folkersen
- Department of Medicine and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Rona J Strawbridge
- Department of Medicine and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway Institute of Clinical Medicine, University of Oslo, Norway
| | - Arne Yndestad
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway K.G. Jebsen Inflammatory Research Center, University of Oslo, Norway
| | - Trine Ranheim
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway Institute of Clinical Medicine, University of Oslo, Norway
| | - Kirsten Krohg-Sørensen
- Department of Thoracic and Cardiovascular Surgery, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Mona Skjelland
- Department of Neurology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Terje Espevik
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway Institute of Clinical Medicine, University of Oslo, Norway K.G. Jebsen Inflammatory Research Center, University of Oslo, Norway
| | - Mariette Lengquist
- Department of Surgery, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Ulf Hedin
- Department of Surgery, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Jan-Håkan Jansson
- Department of Internal Medicine, Skellefteå Hospital and Umeå University Hospital, Umeå, Sweden
| | - Karin Fransén
- Cardiovascular Research Centre, Faculty of Medicine and Health, School of Health and Medical Sciences, Örebro University, Örebro, Sweden
| | - Göran K Hansson
- Department of Medicine and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Per Eriksson
- Department of Medicine and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Allan Sirsjö
- Cardiovascular Research Centre, Faculty of Medicine and Health, School of Health and Medical Sciences, Örebro University, Örebro, Sweden
| |
Collapse
|
18
|
McKie EA, Reid JL, Mistry PC, DeWall SL, Abberley L, Ambery PD, Gil-Extremera B. A Study to Investigate the Efficacy and Safety of an Anti-Interleukin-18 Monoclonal Antibody in the Treatment of Type 2 Diabetes Mellitus. PLoS One 2016; 11:e0150018. [PMID: 26930607 PMCID: PMC4773233 DOI: 10.1371/journal.pone.0150018] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 02/08/2016] [Indexed: 11/23/2022] Open
Abstract
Objective Evidence suggests that chronic subclinical inflammation plays an important role in the pathogenesis of type 2 diabetes (T2DM). Circulating levels of interleukin (IL)-18 appear to be associated with a number of micro- and macrovascular comorbidities of obesity and T2DM. This study was designed to investigate whether inhibition of IL-18 had any therapeutic benefit in the treatment of T2DM. Preliminary efficacy, safety and tolerability, pharmacokinetics, and pharmacodynamics of the anti-IL-18 monoclonal antibody, GSK1070806, were assessed. Research Design and Methods This was a multicentre, randomized, single-blind (sponsor-unblinded), placebo-controlled, parallel-group, phase IIa trial. Obese patients of either sex, aged 18–70 years, with poorly controlled T2DM on metformin monotherapy were recruited. Patients received two doses, of placebo (n = 12), GSK1070806 0.25 mg/kg (n = 13) or GSK1070806 5 mg/kg (n = 12). The primary end-point was the change from baseline in fasting plasma glucose and weighted mean glucose area under the curve (AUC)(0–4 hours) postmixed meal test on Days 29, 57, and 85. Results Thirty-seven patients were randomized to one of the three treatment arms. There were no statistically significant effects of GSK1070806 doses on fasting plasma glucose levels, or weighted mean glucose AUC(0–4 hours) compared with placebo. Conclusions GSK1070806 was well tolerated, and inhibition of IL-18 did not lead to any improvements in glucose control. However, because of study limitations, smaller, potentially clinically meaningful effects of IL-18 inhibition cannot be excluded. Trial Registration ClinicalTrials.gov NCT01648153
Collapse
Affiliation(s)
| | - Juliet L. Reid
- Immunoinflammation Therapy Area Unit, GlaxoSmithKline, Stevenage, United Kingdom
| | | | - Stephen L. DeWall
- Clinical Immunology, Biopharm R&D, GlaxoSmithKline, King of Prussia, Pennsylvania, United States of America
| | - Lee Abberley
- Drug Metabolism and Pharmacokinetics, GlaxoSmithKline, King of Prussia, Pennsylvania, United States of America
| | - Philip D. Ambery
- GlaxoSmithKline, Clinical development, Cardiovascular and Metabolic Medicines Development Centre, London, United Kingdom
| | | |
Collapse
|
19
|
Usefulness of serum interleukin-18 in predicting cardiovascular mortality in patients with chronic kidney disease--systems and clinical approach. Sci Rep 2015; 5:18332. [PMID: 26669254 PMCID: PMC4680867 DOI: 10.1038/srep18332] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 11/16/2015] [Indexed: 01/06/2023] Open
Abstract
The aim of this study was to check if serum interleukin-18 (IL-18) predicts 2-year cardiovascular mortality in patients at various stages of chronic kidney disease (CKD) and history of acute myocardial infarction (AMI) within the previous year. Diabetes mellitus was one of the key factors of exclusion. It was found that an increase in serum concentration of IL-18 above the cut-off point (1584.5 pg/mL) was characterized by 20.63-fold higher risk of cardiovascular deaths among studied patients. IL-18 serum concentration was found to be superior to the well-known cardiovascular risk parameters, like high sensitivity C-reactive protein (hsCRP), carotid intima media thickness (CIMT), glomerular filtration rate, albumins, ferritin, N-terminal prohormone of brain natriuretic peptide (NT-proBNP) in prognosis of cardiovascular mortality. The best predictive for IL-18 were 4 variables, such as CIMT, NT-proBNP, albumins and hsCRP, as they predicted its concentration at 89.5%. Concluding, IL-18 seems to be important indicator and predictor of cardiovascular death in two-year follow-up among non-diabetic patients suffering from CKD, with history of AMI in the previous year. The importance of IL-18 in the process of atherosclerotic plaque formation has been confirmed by systems analysis based on a formal model expressed in the language of Petri nets theory.
Collapse
|
20
|
Deser SB, Bayoglu B, Besirli K, Cengiz M, Arapi B, Junusbekov Y, Dirican A, Arslan C. Increased IL18 mRNA levels in peripheral artery disease and its association with triglyceride and LDL cholesterol levels: a pilot study. Heart Vessels 2015; 31:976-84. [PMID: 26438531 DOI: 10.1007/s00380-015-0753-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 09/25/2015] [Indexed: 11/30/2022]
Abstract
Peripheral artery disease (PAD) typically refers to lower limb vessel ischemia caused by atherosclerotic stenosis of lower extremity arteries. IL18 is a pleiotropic pro-inflammatory cytokine reported to function as an inflammatory biomarker in cardiovascular diseases. IL18 activity is balanced by high-affinity naturally occurring IL18-binding protein (IL18BP). This study aimed to determine whether IL18, IL18 BP mRNA levels and -137 G/C (rs187238) polymorphism, which was previously associated with IL18 gene transcriptional activity, were associated with PAD etiology. IL18, IL18BP mRNA levels from peripheral blood mononuclear cells and -137 G/C (rs187238) polymorphism were determined by quantitative real-time polymerase chain reaction (qRT-PCR) and RT-PCR, respectively, in 55 PAD patients (26 aorta-iliac, 29 femoro-popliteal) and 61 disease-free controls. IL18 mRNA levels were increased in PAD patients compared with healthy controls (p = 0.09); however, did not reach a statistical significant level, also did not significantly differ between aorta-iliac and femoro-popliteal occlusive PAD subgroups (p = 0.285). However, IL18BP mRNA levels were significantly lower in PAD group compared with controls (p < 0.001). Genotype frequencies of rs187238 polymorphism did not significantly differ between PAD patients and controls (p = 0.385). IL18 mRNA levels were significantly correlated with triglycerides and LDL cholesterol levels in PAD patients (p = 0.003, p = 0.014, respectively). HDL cholesterol levels were negatively correlated with IL18 mRNA levels in controls (p = 0.05). This report is a preliminary study to show an association between IL18, IL18BP mRNA levels and PAD and suggests that the IL18 gene may have a significant relationship with triglyceride and LDL cholesterol levels in PAD patients.
Collapse
Affiliation(s)
- Serkan Burc Deser
- Department of Cardiovascular Surgery, Medical Faculty, 19 Mayis University, Samsun, 55139, Turkey.
| | - Burcu Bayoglu
- Department of Medical Biology, Cerrahpasa Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Kazım Besirli
- Department of Cardiovascular Surgery, Cerrahpasa Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Mujgan Cengiz
- Department of Medical Biology, Cerrahpasa Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Berk Arapi
- Department of Cardiovascular Surgery, Cerrahpasa Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Yerik Junusbekov
- Department of Cardiovascular Surgery, Cerrahpasa Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Ahmet Dirican
- Department of Biostatistics and Medical Informatics, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Caner Arslan
- Department of Cardiovascular Surgery, Cerrahpasa Medical Faculty, Istanbul University, Istanbul, Turkey
| |
Collapse
|
21
|
Wang J, Sun C, Gerdes N, Liu C, Liao M, Liu J, Shi MA, He A, Zhou Y, Sukhova GK, Chen H, Cheng XW, Kuzuya M, Murohara T, Zhang J, Cheng X, Jiang M, Shull GE, Rogers S, Yang CL, Ke Q, Jelen S, Bindels R, Ellison DH, Jarolim P, Libby P, Shi GP. Interleukin 18 function in atherosclerosis is mediated by the interleukin 18 receptor and the Na-Cl co-transporter. Nat Med 2015; 21:820-6. [PMID: 26099046 PMCID: PMC4554539 DOI: 10.1038/nm.3890] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 05/26/2015] [Indexed: 12/13/2022]
Abstract
Interleukin-18 (IL18) participates in atherogenesis through several putative mechanisms. Interruption of IL18 action reduces atherosclerosis in mice. Here, we show that absence of the IL18 receptor (IL18r) does not affect atherosclerosis in apolipoprotein E-deficient (Apoe(-/-)) mice, nor does it affect IL18 cell surface binding to or signaling in endothelial cells. As identified initially by co-immunoprecipitation with IL18, we found that IL18 interacts with the Na-Cl co-transporter (NCC; also known as SLC12A3), a 12-transmembrane-domain ion transporter protein preferentially expressed in the kidney. NCC is expressed in atherosclerotic lesions, where it colocalizes with IL18r. In Apoe(-/-) mice, combined deficiency of IL18r and NCC, but not single deficiency of either protein, protects mice from atherosclerosis. Peritoneal macrophages from Apoe(-/-) mice or from Apoe(-/-) mice lacking IL18r or NCC show IL18 binding and induction of cell signaling and cytokine and chemokine expression, but macrophages from Apoe(-/-) mice with combined deficiency of IL18r and NCC have a blunted response. An interaction between NCC and IL18r on macrophages was detected by co-immunoprecipitation. IL18 binds to the cell surface of NCC-transfected COS-7 cells, which do not express IL18r, and induces cell signaling and cytokine expression. This study identifies NCC as an IL18-binding protein that collaborates with IL18r in cell signaling, inflammatory molecule expression, and experimental atherogenesis.
Collapse
Affiliation(s)
- Jing Wang
- 1] Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA. [2] State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Tsinghua University, Beijing, China
| | - Chongxiu Sun
- 1] Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA. [2] Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, China
| | - Norbert Gerdes
- 1] Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA. [2] Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians University Munich, Munich, Germany
| | - Conglin Liu
- 1] Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA. [2] Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengyang Liao
- 1] Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA. [2] Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Jian Liu
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Michael A Shi
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Aina He
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Yi Zhou
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Galina K Sukhova
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Huimei Chen
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Xian Wu Cheng
- Departments of Cardiology and Geriatrics, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Masafumi Kuzuya
- Departments of Cardiology and Geriatrics, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Toyoaki Murohara
- Departments of Cardiology and Geriatrics, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Jie Zhang
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Xiang Cheng
- 1] Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA. [2] Institute of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Mengmeng Jiang
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Gary E Shull
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Shaunessy Rogers
- Division of Nephrology and Hypertension, Oregon Health and Science University, Portland, Oregon, USA
| | - Chao-Ling Yang
- Division of Nephrology and Hypertension, Oregon Health and Science University, Portland, Oregon, USA
| | - Qiang Ke
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Sabina Jelen
- Department of Physiology, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | - René Bindels
- Department of Physiology, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | - David H Ellison
- Division of Nephrology and Hypertension, Oregon Health and Science University, Portland, Oregon, USA
| | - Petr Jarolim
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Peter Libby
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
22
|
Xie SL, Chen YY, Zhang HF, Deng BQ, Shu XR, Su ZZ, Lin YQ, Nie RQ, Wang JF. Interleukin 18 and extracellular matrix metalloproteinase inducer cross-regulation: implications in acute myocardial infarction. Transl Res 2015; 165:387-95. [PMID: 25267095 DOI: 10.1016/j.trsl.2014.09.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 08/30/2014] [Accepted: 09/04/2014] [Indexed: 01/02/2023]
Abstract
Circulating interleukin-18 (IL-18) is thought to promote atherosclerosis and cardiovascular complications such as plaque rupture. Atherosclerosis is also characterized by smooth muscle cell migration, a consequence of extracellular matrix (ECM) degradation regulated by metalloproteinases (MMPs). Because extracellular matrix metalloproteinase inducer (EMMPRIN) has been shown to promote plaque instability by inducing ECM degradation and MMP synthesis, we investigated whether a cross-regulatory interaction exists between IL-18 and EMMPRIN in human monocytes. EMMPRIN levels in monocytes were markedly greater in 20 patients with acute myocardial infarction (AMI) compared with 20 patients with stable angina pectoris or 20 healthy volunteers (control group). The levels of IL-18 and MMP-9 in serum were also significantly greater in the AMI group in comparison with the other 2 groups. IL-18 levels positively correlated with increased levels of EMMPRIN in monocytes. In vitro, the expression of EMMPRIN was increased in monocytes cultured with IL-18, and IL-18 secretion was augmented in monocytes cultured with EMMPRIN. Gene silencing of EMMPRIN by small interfering RNA reduced monocyte secretion of both IL-18 and MMP-9. In the present study, cross-regulation between IL-18 and EMMPRIN in monocytes was demonstrated. This interaction may amplify the inflammatory cascade and be responsible for increased monocytic MMP-9 serum levels in atherosclerosis, contributing to atherosclerotic plaque destabilization and subsequent AMI.
Collapse
Affiliation(s)
- Shuang-Lun Xie
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China
| | - Yu-Yang Chen
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China
| | - Hai-Feng Zhang
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China
| | - Bing-Qing Deng
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China
| | - Xiao-Rong Shu
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China
| | - Zi-Zhuo Su
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China
| | - Yong-Qing Lin
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China
| | - Ru-Qiong Nie
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China
| | - Jing-Feng Wang
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China.
| |
Collapse
|
23
|
McLeod O, Dunér P, Samnegård A, Tornvall P, Nilsson J, Hamsten A, Bengtsson E. Autoantibodies against basement membrane collagen type IV are associated with myocardial infarction. IJC HEART & VASCULATURE 2014; 6:42-47. [PMID: 28785625 PMCID: PMC5497157 DOI: 10.1016/j.ijcha.2014.12.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 12/20/2014] [Indexed: 12/03/2022]
Abstract
Background Collagen type IV is the major constituent of basement membranes underlying endothelial cells and is important for endothelial cell attachment and function. Autoantibodies against native collagen type IV have been found in various autoimmune diseases. Oxidation of LDL in the vascular wall results in the formation of reactive aldehydes, which could modify surrounding matrix proteins. Like oxidized LDL, these modified matrix proteins are likely to induce immune responses. We examined whether autoantibodies against native or aldehyde-modified collagen type IV are associated with myocardial infarction. Methods IgM and IgG against native and aldehyde-modified collagen type IV were measured by ELISA in serum from 387 survivors of a first myocardial infarction and 387 age- and sex-matched controls. Results Post-infarction patients had significantly increased levels of IgM against native collagen type IV, and IgG against native collagen type IV was present at detectable level in 17% of patients as opposed to 7% of controls (p < 0.001). Controlling for major cardiovascular risk factors demonstrated that the presence of IgG against native collagen type IV was associated with myocardial infarction (OR 2.9 (1.6–5.4), p = 0.001). Similarly, subjects in the highest quartile of IgM against native collagen type IV had increased risk of having suffered myocardial infarction (OR 3.11 (1.8–5.4), p < 0.001) after adjusting for cardiovascular risk factors. In contrast, IgG against aldehyde-modified collagen type IV was decreased in myocardial infarction patients, but this association was not independent of established cardiovascular risk factors. Conclusion Autoantibodies against collagen type IV are associated with myocardial infarction independently of traditional cardiovascular risk factors. We measured native and MDA-collagen type IV IgM and IgG in MI patients and controls. Post-infarction patients had increased levels of IgM against native collagen type IV. Presence of IgG against native collagen type IV was associated with MI. In contrast, IgG against MDA-collagen type IV was decreased in MI patients.
Collapse
Affiliation(s)
- Olga McLeod
- Department of Medicine, Atherosclerosis Research Unit, Karolinska Institutet, Stockholm, Sweden
| | - Pontus Dunér
- Department of Clinical Sciences, Skåne University Hospital, Malmö, Sweden
| | - Ann Samnegård
- Department of Clinical Sciences, Cardiology Unit, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Per Tornvall
- Department of Clinical Sciences, Cardiology Unit, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Jan Nilsson
- Department of Clinical Sciences, Skåne University Hospital, Malmö, Sweden
| | - Anders Hamsten
- Department of Medicine, Atherosclerosis Research Unit, Karolinska Institutet, Stockholm, Sweden
| | - Eva Bengtsson
- Department of Clinical Sciences, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
24
|
Bhat OM, Kumar PU, Giridharan NV, Kaul D, Kumar MJM, Dhawan V. Interleukin-18-induced atherosclerosis involves CD36 and NF-κB crosstalk in Apo E-/- mice. J Cardiol 2014; 66:28-35. [PMID: 25475966 DOI: 10.1016/j.jjcc.2014.10.012] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 09/15/2014] [Accepted: 10/06/2014] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Interleukin (IL)-18 is a pleotropic cytokine involved in various inflammatory disorders. The transcription factor, nuclear factor kappa-B (NF-κB), is thought to play an important role in IL-18 signaling. The present study proposes a novel role for IL-18 in cholesterol efflux and plaque stability and demonstrates that pyrrolidine dithiocarbamate (PDTC), a NF-κB inhibitor blocks IL-18 signaling in apolipoprotein (Apo) E-/- mice. METHODS Three groups of normal chow-diet-fed, male Apo E-/- mice, aged 12 weeks (n=6/group) were employed: Gp I, PBS (2mo); Gp II, recombinant (r)IL-18 (1mo) followed by PBS (1mo); Gp III, rIL-18 (1mo) followed by PDTC (1mo). RESULTS Significantly augmented expression of IL-18 receptor (R)α by fluorescence-activated cell sorting analysis and plasma IL-18 was observed in Gp II. There was a significant increase in total cholesterol and low-density lipoprotein cholesterol whereas high-density lipoprotein cholesterol was significantly decreased in Gp II. However, this pattern was reversed in Gp III. Significantly augmented mRNA expression of IL-18, CD36, matrix metalloproteinase (MMP)-9, and NF-κB was observed in Gp II but liver X receptor alpha (LXR-α) gene was significantly reduced. A significant increase in frequency of atherosclerotic lesions was observed in Gp II animals, whereas there was a significant decrease in the Gp III. CONCLUSION IL-18 administration initiates inflammatory cascade by binding with IL-18 Rα via NF-κB which is involved in progression and destabilization of atherosclerotic plaques in Apo E-/- mice. This study also reveals that NF-κB blockade with PDTC, blocks IL-18 signaling through down-regulation of IL-18, IL-18 Rα, CD36, and MMP-9, thus reducing inflammation and restoring plaque instability via upregulation of LXR-α.
Collapse
Affiliation(s)
- Owais Mohammad Bhat
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - P Uday Kumar
- National Centre for Laboratory Animal Sciences (NCLAS), Hyderabad, Department of Histopathology, National Institute of Nutrition (NIN), Hyderabad, India
| | - N V Giridharan
- Amrita School of Nanosciences & Molecular Medicine, Amrita Institute of Medical Sciences, Amrita Viswavidyapeedham, Kochi, Kerala, India
| | - Deepak Kaul
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - M J Mahesh Kumar
- Animal House, Centre for Cellular and Molecular Biology (CCMB), Hyderabad, India
| | - Veena Dhawan
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India.
| |
Collapse
|
25
|
Battes LC, Cheng JM, Oemrawsingh RM, Boersma E, Garcia-Garcia HM, de Boer SPM, Buljubasic N, Mieghem NAV, Regar E, Geuns RJV, Serruys PW, Akkerhuis KM, Kardys I. Circulating cytokines in relation to the extent and composition of coronary atherosclerosis: results from the ATHEROREMO-IVUS study. Atherosclerosis 2014; 236:18-24. [PMID: 25003948 DOI: 10.1016/j.atherosclerosis.2014.06.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 06/15/2014] [Accepted: 06/16/2014] [Indexed: 01/07/2023]
Abstract
OBJECTIVE We investigated whether concentrations of TNF-α, TNF-β, TNF-receptor 2, interferon-γ, IL-6, IL-8, IL-10 and IL-18 are associated with extent and composition of coronary atherosclerosis determined by grayscale and virtual histology (VH)- intravascular ultrasound (IVUS). METHODS Between 2008 and 2011, IVUS(-VH) imaging of a non-culprit coronary artery was performed in 581 patients (stable angina pectoris (SAP), n = 261; acute coronary syndrome (ACS), n = 309) undergoing coronary angiography from the ATHEROREMO-IVUS study. Plaque burden, presence of VH-IVUS-derived thin-cap fibroatheroma (TCFA) lesions, and presence of VH-TCFA lesions with plaque burden ≥70% were assessed. Blood samples for cytokine measurement were drawn from the arterial sheath prior to the angiography procedure. We applied linear and logistic regression. RESULTS TNF-α levels were positively associated with plaque burden (beta (β) [95%CI]: 4.45 [0.99-7.91], for highest vs lowest TNF-α tertile) and presence of VH-TCFA lesions (odds ratio (OR) [95%CI] 2.30 (1.17-4.52), highest vs lowest TNF-α tertile) in SAP patients. Overall, an inverse association was found between IL-10 concentration and plaque burden (β [95%CI]: -1.52 [-2.49 to -0.55], per Ln (pg/mL) IL-10) as well as IL-10 and VH-TCFA lesions with plaque burden ≥70% (OR: 0.31 [0.12-0.80],highest vs lowest IL-10 tertile). These effects did not reach statistical significance in the separate SAP and ACS groups. CONCLUSION Higher circulating TNF-α was associated with higher plaque burden and VH-TCFA lesions in SAP patients. Lower circulating IL-10 was associated with higher plaque burden and large VH-TCFA lesions. These in-vivo findings suggest a role for these cytokines in extent and vulnerability of atherosclerosis.
Collapse
Affiliation(s)
- Linda C Battes
- Clinical Epidemiology Unit, Department of Cardiology, Erasmus MC, Thoraxcenter, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands.
| | - Jin M Cheng
- Clinical Epidemiology Unit, Department of Cardiology, Erasmus MC, Thoraxcenter, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands.
| | - Rohit M Oemrawsingh
- Clinical Epidemiology Unit, Department of Cardiology, Erasmus MC, Thoraxcenter, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands.
| | - Eric Boersma
- Clinical Epidemiology Unit, Department of Cardiology, Erasmus MC, Thoraxcenter, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Hector M Garcia-Garcia
- Clinical Epidemiology Unit, Department of Cardiology, Erasmus MC, Thoraxcenter, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands.
| | - Sanneke P M de Boer
- Clinical Epidemiology Unit, Department of Cardiology, Erasmus MC, Thoraxcenter, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands.
| | - Nermina Buljubasic
- Clinical Epidemiology Unit, Department of Cardiology, Erasmus MC, Thoraxcenter, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands.
| | - Nicolas A van Mieghem
- Clinical Epidemiology Unit, Department of Cardiology, Erasmus MC, Thoraxcenter, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands.
| | - Evelyn Regar
- Clinical Epidemiology Unit, Department of Cardiology, Erasmus MC, Thoraxcenter, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands.
| | - Robert-Jan van Geuns
- Clinical Epidemiology Unit, Department of Cardiology, Erasmus MC, Thoraxcenter, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands.
| | - Patrick W Serruys
- Clinical Epidemiology Unit, Department of Cardiology, Erasmus MC, Thoraxcenter, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands.
| | - K Martijn Akkerhuis
- Clinical Epidemiology Unit, Department of Cardiology, Erasmus MC, Thoraxcenter, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands.
| | - Isabella Kardys
- Clinical Epidemiology Unit, Department of Cardiology, Erasmus MC, Thoraxcenter, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands.
| |
Collapse
|
26
|
Plasma granzyme B in ST elevation myocardial infarction versus non-ST elevation acute coronary syndrome: comparisons with IL-18 and fractalkine. Mediators Inflamm 2013; 2013:343268. [PMID: 24307760 PMCID: PMC3836447 DOI: 10.1155/2013/343268] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 09/12/2013] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE The proapoptotic protein, granzyme B (GZB), was identified as a contributor to the atherosclerotic plaque instability and recently as inflammatory activator. We studied the release kinetics of GZB and other markers of inflammation such as high sensitivity C reactive protein (hsCRP), interleukin 18 (IL-18), and fractalkine (FKN) in the early phase after acute cardiac events in different ACS subgroups. METHODS Thirty-six nondiabetic patients with ACS were compared to 12 control subjects. According to ACS diagnosis, the patients were classified into 22 patients with ST elevation myocardial infarction (STEMI) and 14 patients with non-ST elevation myocardial infarction or unstable angina (NSTEMI/UA). Blood samples were taken on day 1 (day of onset) and day 3 to measure hsCRP, IL-18, FKN, and GZB by ELISA. RESULTS Patients with ACS showed significantly higher GZB, IL-18, and FKN levels than the controls. STEMI group showed significantly higher GZB levels than NSTEMI/UA group. On day 3, FKN levels displayed a significant decrease, while GZB levels were significantly increased. IL-18 levels were more or less constant. GZB levels were positively correlated with IL-18 (r = 0.416, P < 0.01) and FKN (r = 0.58, P < 0.001). CONCLUSIONS Unlike IL-18 and FKN, plasma GZB may be a marker of ACS disease severity.
Collapse
|
27
|
Interleukin-18 Promoter Gene Polymorphisms are not Associated with Myocardial Infarction in Type 2 Diabetes in Slovenia. Balkan J Med Genet 2013; 14:3-9. [PMID: 24052696 PMCID: PMC3776693 DOI: 10.2478/v10034-011-0011-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Type 2 diabetes is a major risk factor for myocardial infarction (MI) and chronic inflammation may play a central role in both diseases. Interleukin (IL)-18 is a potent proinflammatory cytokine, which is considered important in acute coronary syndromes and type 2 diabetes. We investigated the association of the −137 (G>C), polymorphism (rs187238) and the −607 (C>A) polymorphism (rs1946518) of the IL-18 gene promoter region in 495 Caucasians with type 2 diabetes, of whom 169 had MI and 326 subjects had no clinically evident coronary artery disease (controls). We also investigated the impact of these polymorphisms on the serum IL-18 level in subsets of both groups and in a normal group. Genotype distributions of the polymorphisms showed no significant difference between cases and controls. However, IL-18 serum levels were significantly lower in diabetics with the 137 CC genotype than in those with other genotypes (241.5 ± 132.7 ng/L vs. 340.2 ± 167.4 ng/L; p <0.05). High sensitivity C-reactive protein and IL-18 serum levels were higher in diabetics in the MI group than in the control group. We conclude that these IL-18 promoter gene polymorphisms are not risk factors for MI in Caucasians with type 2 diabetes.
Collapse
|
28
|
CARD8 gene encoding a protein of innate immunity is expressed in human atherosclerosis and associated with markers of inflammation. Clin Sci (Lond) 2013; 125:401-7. [PMID: 23611467 DOI: 10.1042/cs20120572] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Inflammation is a key factor in the development of atherosclerotic coronary artery disease. It is promoted through the inflammasome, a molecular machine that produces IL (interleukin)-1β in response to cholesterol crystal accumulation in macrophages. The CARD8 (caspase recruitment domain 8) protein modulates this process by suppressing caspase 1 and the transcription factor NF-κB (nuclear factor κB). The expression of CARD8 mRNA was examined in atherosclerotic vascular tissue and the impact on MI (myocardial infarction) of a polymorphism in the CARD8 gene determined. CARD8 mRNA was analysed by microarray of human atherosclerotic tissue and compared with transplant donor arterial tissue. Microarray analysis was performed for proximal genes associated with the rs2043211 locus in plaque. The CARD8 rs2043211 polymorphism was analysed by genotyping of two Swedish MI cohorts, FIA (First Myocardial Infarction in Northern Sweden) and SCARF (Stockholm Coronary Atherosclerosis Risk Factor). The CRP (C-reactive protein) level was measured in both cohorts, but the levels of the pro-inflammatory cytokines IL-1β, IL-18, TNF (tumour necrosis factor) and MCP-1 (monocyte chemoattractant protein) were measured in sera available from the SCARF cohort. CARD8 mRNA was highly expressed in atherosclerotic plaques compared with the expression in transplant donor vessel (P<0.00001). The minor allele was associated with lower expression of CARD8 in the plaques, suggesting that CARD8 may promote inflammation. Carriers of the minor allele of the rs2043211 polymorphism also displayed lower circulating CRP and lower levels of the pro-atherosclerotic chemokine MCP-1. However, no significant association could be detected between this polymorphism and MI in the two cohorts. Genetic alterations in the CARD8 gene therefore seem to be of limited importance for the development of MI.
Collapse
|
29
|
Tajik N, Keshavarz SA, Masoudkabir F, Djalali M, Sadrzadeh-Yeganeh HH, Eshraghian MR, Chamary M, Ahmadivand Z, Yazdani T, Javanbakht MH. Effect of diet-induced weight loss on inflammatory cytokines in obese women. J Endocrinol Invest 2013; 36:211-5. [PMID: 22733212 DOI: 10.3275/8465] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Obesity is associated with lowgrade systemic inflammation which has been linked to the increased risk of cardiovascular disease and Type 2 diabetes in obese patients. AIM To evaluate changes in pro/anti-inflammatory adipocytokines and metabolic profile after moderate diet-induced weight loss. SUBJECTS AND METHODS Twenty-nine pre-menopausal obese women (body mass index ≥30 kg/m2) aged 21 to 54 years without diabetes, hypertension, or hyperlipidemia, were enrolled in this study. We measured anthropometric parameters, lipid and glucose profiles, interleukin (IL)-6, IL-10, and IL-18 in obese women, who then entered a medically supervised program aimed at reducing body weight by 10% or more. Obese women restricted their caloric intake (by 500-1000 kcal/day) and consumed 50 g/day of a fiber supplement (Slim Last Powder) for 12 weeks. RESULTS By completing the dietary intervention program, weight (Δ = -10.0%, p<0.0001), body mass index, waist circumference, triceps skinfold thickness, total cholesterol, triglyceride, and fasting plasma glucose significantly decreased, while HDL-cholesterol significantly increased. While plasma levels of IL-6 and IL-18 decreased by 27% after 12 weeks, no significant change was observed in circulating levels of IL-10. CONCLUSION Our study suggests that an improved body composition induced by restriction of energy intake is associated with favorable serum concentrations of IL-6 and IL-18 in obese women. However, the anti-inflammatory IL-10 is not affected by a moderate weight decrease.
Collapse
Affiliation(s)
- N Tajik
- Nutrition and Biochemistry Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Justina Farias TD, do Canto LM, Delagnelo Medeiros M, Rodrigues Sereia AF, de Carlos Back LKF, de Mello FM, Fontes Zimmermann A, Alves Pereira I, Netto Muniz YC, Marrero AR, de Souza IR. Ausência de associação entre os polimorfismos do gene interleucina-18 e artrite reumatoide. REVISTA BRASILEIRA DE REUMATOLOGIA 2013. [DOI: 10.1590/s0482-50042013000200007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
|
31
|
Inflammatory biomarkers CRP, MCP-1, serum amyloid alpha and interleukin-18 in patients with HTN and dyslipidemia: impact of diabetes mellitus on metabolic syndrome and the effect of statin therapy. Hypertens Res 2013; 36:550-8. [DOI: 10.1038/hr.2012.214] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
32
|
Hong K, Oh K, Lee S, Hong J, Choi J, Kwak A, Kang D, Kim E, Jo S, Jhun H, Kim S. Recombinant Fc-IL-18BPc Isoform Inhibits IL-18-Induced Cytokine Production. Hybridoma (Larchmt) 2012; 31:99-104. [DOI: 10.1089/hyb.2011.0085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Kwangwon Hong
- Laboratory of Cytokine Immunology, Department of Biomedical Sciences and Technology, Konkuk University Medical Center, School of Medicine, Konkuk University, Seoul, Korea
| | - Kwangjun Oh
- Laboratory of Cytokine Immunology, Department of Biomedical Sciences and Technology, Konkuk University Medical Center, School of Medicine, Konkuk University, Seoul, Korea
- Department of Orthopaedic Surgery, Konkuk University Medical Center, School of Medicine, Konkuk University, Seoul, Korea
| | - Siyoung Lee
- Laboratory of Cytokine Immunology, Department of Biomedical Sciences and Technology, Konkuk University Medical Center, School of Medicine, Konkuk University, Seoul, Korea
| | - Jaewoo Hong
- Laboratory of Cytokine Immunology, Department of Biomedical Sciences and Technology, Konkuk University Medical Center, School of Medicine, Konkuk University, Seoul, Korea
| | - Jida Choi
- Laboratory of Cytokine Immunology, Department of Biomedical Sciences and Technology, Konkuk University Medical Center, School of Medicine, Konkuk University, Seoul, Korea
| | - Areum Kwak
- Laboratory of Cytokine Immunology, Department of Biomedical Sciences and Technology, Konkuk University Medical Center, School of Medicine, Konkuk University, Seoul, Korea
| | - Dongjun Kang
- Laboratory of Cytokine Immunology, Department of Biomedical Sciences and Technology, Konkuk University Medical Center, School of Medicine, Konkuk University, Seoul, Korea
| | - Eunsom Kim
- Laboratory of Cytokine Immunology, Department of Biomedical Sciences and Technology, Konkuk University Medical Center, School of Medicine, Konkuk University, Seoul, Korea
| | - Seunghyun Jo
- Laboratory of Cytokine Immunology, Department of Biomedical Sciences and Technology, Konkuk University Medical Center, School of Medicine, Konkuk University, Seoul, Korea
| | - Hyunjhung Jhun
- Laboratory of Cytokine Immunology, Department of Biomedical Sciences and Technology, Konkuk University Medical Center, School of Medicine, Konkuk University, Seoul, Korea
| | - Soohyun Kim
- Laboratory of Cytokine Immunology, Department of Biomedical Sciences and Technology, Konkuk University Medical Center, School of Medicine, Konkuk University, Seoul, Korea
| |
Collapse
|
33
|
Chai HT, Chen YL, Chung SY, Tsai TH, Yang CH, Chen HC, Sung PH, Sun CK, Chang LT, Fan CQ, Yip HK. Value and level of plasma homocysteine in patients with angina pectoris undergoing coronary angiographic study. Int Heart J 2011; 52:280-5. [PMID: 22008436 DOI: 10.1536/ihj.52.280] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
This study tested whether the plasma level of total homocysteine (tHcy) was predictive of obstructive coronary artery disease (CAD) and clinical outcome in patients undergoing coronary angiographic (CAG) study. From September 2002 to October 2004, 1,305 consecutive patients with angina pectoris undergoing CAG study were consecutively enrolled. Blood samples were prospectively collected to assess the plasma level of tHcy from each patient before catheterization. Of these 1305 patients, 676 (51.8%) had multivessel disease (group 1), 367 (28.1%) had single-vessel disease (group 2), and 262 (20.1%) had normal coronary artery or insignificant coronary artery disease (group 3). The plasma level of tHcy was notably higher in group 1 than in groups 2 and 3 (11.6 ± 4.4 versus 10.9 ± 4.0 versus 10.4 ± 3.8, P < 0.001). Univariate binary logistic regression analysis demonstrated that the plasma tHcy level was strongly associated with multiple-vessel disease (MVD) (defined as ≥ 2 vessel disease) (P < 0.001). Multivariate binary logistic regression analysis showed that tHcy level, fasting blood sugar, diabetes mellitus, and age were significantly and independently predictive of MVD (all P < 0.03). Univariate Cox regression analysis demonstrated that tHcy level was predictive of long-term mortality (P = 0.042). However, the tHcy level was not an independent predictor of long-term mortality on multivariate Cox regression analysis (P > 0.05). The results of our study support the hypothesis that tHcy level is an independent predictor of MVD in patients with chest pain undergoing CAG study. Conversely, our study did not support the tHcy level as an independent predictor of long-term mortality in this clinical setting.
Collapse
Affiliation(s)
- Han-Tan Chai
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan, ROC
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Marcondes MCG, Zhukov V, Bradlow H, Sanchez-Alavez M, Gonzalez ASA, Curtiss LK, Conti B. Effects of chronic mental stress and atherogenic diet on the immune inflammatory environment in mouse aorta. Brain Behav Immun 2011; 25:1649-57. [PMID: 21722726 PMCID: PMC3191305 DOI: 10.1016/j.bbi.2011.06.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 06/07/2011] [Accepted: 06/10/2011] [Indexed: 11/18/2022] Open
Abstract
Inflammation and stress are regarded as two important atherogenic factors. Because stress can affect leukocyte distribution, we hypothesized that stress-mediated leukocyte extravasation can modify the inflammatory environment of the arterial wall possibly contributing to atherogenesis. To test this hypothesis we evaluated the inflammatory environment of the aorta in C57Bl/6 mice subjected to 3 and 12 months of chronic stress and compared it to age matched non-stressed animals. Experiments were carried out in mice fed regular chow or atherogenic diets. Both treatments increased the expression of vascular and leukocyte adhesion molecules and leukocyte accumulation. At 3 months, stress but not an atherogenic diet elevated the number of CD4 cells, CD8 cells, macrophages, dendritic cells and neutrophils. These changes were associated with elevation of transcripts for ICAM-1 and VCAM-1, E-selectin and neuropeptide Y. At 12 months, stress or high cholesterol acted similarly to elevate the number of CD8 and macrophages, and synergistically on the number of all cell types investigated. At this time-point, strong synergism was also observed on the level of E-selectin and NPY in the aorta, but not in the circulation. Despite these effects, histological and morphological alterations of the arterial wall were severe in the atherogenic diet, but not in the stress groups. Thus, although stress and an atherogenic diet may both affect leukocyte accumulation in the aorta, they may contribute differently to atherogenesis.
Collapse
|
35
|
Christersson C, Oldgren J, Wallentin L, Siegbahn A. Treatment with an oral direct thrombin inhibitor decreases platelet activity but increases markers of inflammation in patients with myocardial infarction. J Intern Med 2011; 270:215-23. [PMID: 21255134 DOI: 10.1111/j.1365-2796.2011.02354.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Thrombin has a role not only in the coagulation process but also in inflammatory responses. Oral direct thrombin inhibitors (DTIs) are currently being evaluated in patients with thromboembolic diseases. OBJECTIVE To investigate whether an oral DTI affects markers for platelet and inflammatory activity after myocardial infarction (MI). METHODS A total of 518 patients with MI were randomly assigned to ximelagatran treatment (four different dose groups) in combination with aspirin, or aspirin alone for 6 months. The levels of soluble (s) P-selectin, soluble tissue factor, C-reactive protein (CRP), interleukin (IL)-10 and IL-18 were analysed in serial blood samples. RESULTS sP-selectin concentration increased after 1 week and persisted at an elevated level for 6 months in all study groups (P < 0.001). In the two highest ximelagatran dose groups, there was a reduced increase in sP-selectin compared to treatment with lower doses of ximelagatran and aspirin alone (P = 0.01 and P = 0.002, respectively). IL-18 levels did not change in the aspirin alone treatment group. By contrast, there was an elevation in IL-18 level in the lower and higher ximelagatran dose groups after 6 months (P = 0.006 and P < 0.001, respectively). Ximelagatran increased IL-10 levels (P = 0.002) and reduced the decrease in CRP levels after 6 months compared to treatment with aspirin alone (P = 0.002). CONCLUSION A persistent elevation of platelet activity is found in patients with a recent MI after the cessation of acute antithrombotic treatment, and the addition of an oral DTI at higher doses decreases the activity. By contrast, long-term treatment with a DTI increases the levels of several markers of inflammation. Further studies with prolonged exposure of oral DTIs are needed for evaluation of the effect on inflammatory processes and to determine whether these agents influence clinical outcomes.
Collapse
Affiliation(s)
- C Christersson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
| | | | | | | |
Collapse
|
36
|
Khan DA, Ansari WM, Khan FA. Pro/Anti-Inflammatory Cytokines in the Pathogenesis of Premature Coronary Artery Disease. J Interferon Cytokine Res 2011; 31:561-7. [DOI: 10.1089/jir.2010.0157] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Dilshad Ahmed Khan
- Department of Chemical Pathology and Endocrinology, National University of Science and Technology, Rawalpindi, Pakistan
| | - Wafa Munir Ansari
- Department of Chemical Pathology, Army Medical College, National University of Science and Technology, Rawalpindi, Pakistan
| | - Farooq Ahmed Khan
- Department of Chemical Pathology and Endocrinology, National University of Science and Technology, Rawalpindi, Pakistan
| |
Collapse
|
37
|
Fujioka Y, Fukuda A, Ishida T, Kagimoto S, Nakamura Y, Iwakura A, Hara K, Yamamoto T, Kuroe A, Ohya M, Fujimoto S, Hamamoto Y, Honjo S, Ikeda H, Nabe K, Tsuda K, Taniguchi A, Tanaka K, Koshiyama H, Kume N, Hirata KI. Pitavastatin Reduces Elevated IL-18 Levels in Japanese Subjects with Hypercholesterolemia: Sub-analysis of Kansai Investigation of Statin for Hyperlipidemic Intervention in Metabolism and Endocrinology (KISHIMEN). J Atheroscler Thromb 2011; 18:8-15. [DOI: 10.5551/jat.5942] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
38
|
Fix C, Bingham K, Carver W. Effects of interleukin-18 on cardiac fibroblast function and gene expression. Cytokine 2010; 53:19-28. [PMID: 21050772 DOI: 10.1016/j.cyto.2010.10.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2010] [Revised: 09/10/2010] [Accepted: 10/06/2010] [Indexed: 01/15/2023]
Abstract
Fibroblasts are the primary cell type responsible for synthesis and remodeling of the extracellular matrix in the heart. A number of factors including growth factors, hormones and mechanical forces have been identified that modulate the production of extracellular matrix by cardiac fibroblasts. Inflammatory mediators including pro-inflammatory cytokines and chemokines also impact fibrosis of the heart. Recent studies have illustrated that interleukin-18 promotes a pro-fibrotic response in cardiac fibroblasts; however the effects of this cytokine on other aspects of fibroblast function have not been examined. While fibroblasts have long been known for their role in production and remodeling of the extracellular matrix, other functions of these cells are only now beginning to be appreciated. We hypothesize that exposure to interleukin-18 will stimulate other aspects of fibroblast behavior important in myocardial remodeling including proliferation, migration and collagen reorganization. Fibroblasts were isolated from adult male rat hearts and bioassays performed to determine the effects of interleukin-18 on fibroblast function. Treatment of fibroblasts with interleukin-18 (1-100ng/ml) resulted in increased production of extracellular matrix components and remodeling or contraction of three-dimensional collagen scaffolds by these cells. Furthermore, exposure to interleukin-18 stimulated fibroblast migration and proliferation. Treatment of heart fibroblasts with interleukin-18 resulted in the rapid activation of the c-Jun N-terminal kinase (JNK) and phosphoinositide 3-kinase (PI3-kinase) pathways. Studies with pharmacological inhibitors illustrated that activation of these pathways is critical to interleukin-18 mediated alterations in fibroblast function. These studies illustrate that interleukin-18 plays a role in modulation of cardiac fibroblast function and may be an important component of the inflammation-fibrosis cascade during pathological myocardial remodeling.
Collapse
Affiliation(s)
- Charity Fix
- Department of Cell Biology and Anatomy, University of South Carolina, School of Medicine, Columbia, SC 29209, USA
| | | | | |
Collapse
|
39
|
Hernesniemi JA, Anttila K, Nieminen T, Kähönen M, Mononen N, Nikus K, Turjanmaa V, Viik J, Lehtinen R, Lehtimäki T. IL-18 gene polymorphism, cardiovascular mortality and coronary artery disease. Eur J Clin Invest 2010; 40:994-1001. [PMID: 20735470 DOI: 10.1111/j.1365-2362.2010.02356.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Interleukin 18(IL-18) is a pro-atherosclerotic cytokine. Elevated IL-18 levels and the genetic variation of the IL-18 have been previously linked with acute coronary events and cardiovascular mortality among patients with coronary artery disease (CAD). We studied the possible association between the IL-18 gene polymorphism and cardiovascular mortality during follow-up among Finnish patients who had undergone a clinical exercise stress test, in addition to the possible effect on the expression of angiography-verified CAD. MATERIALS AND METHODS A total of 2152 patients of the Finnish Cardiovascular Study (cohort study) were followed up for 6·3years and cardiovascular mortality was recorded. Angiography was performed on 461 patients. Genotyping of five common single nucleotide polymorphisms (SNPs) of the IL-18 gene was performed using the 5'nuclease assay for allelic discrimination with the ABI Prism 7900HT Sequence Detection System. RESULTS Among the study population, IL-18 gene polymorphism did not associate with cardiovascular mortality. According to adjusted binary regression analysis, the male carriers of one major haplotype (the only ones carrying the t allele of the +127 C/t SNP) had a lower occurrence rate for significant CAD defined as > 50% stenosis in at least one of the main branches of the coronary arteries (OR 0·495, 95% CI 0·862-0·284, P=0·041). No associations were observed among women. The sex-by-genotype interaction was significant (P=0·033). CONCLUSIONS The IL-18 gene was not found to associate significantly with mortality. Among patients who had coronary angiography, one major haplotype of the IL-18 gene has a gender-dependent different impact on the expression of CAD.
Collapse
Affiliation(s)
- Jussi A Hernesniemi
- Laboratory of Atherosclerosis Genetics, Department of Clinical Chemistry, Tampere University Hospital, Centre for Laboratory Medicine, Tampere, Finland Medical School, University of Tampere, Tampere, Finland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Stein S, Lohmann C, Handschin C, Stenfeldt E, Borén J, Lüscher TF, Matter CM. ApoE-/- PGC-1α-/- mice display reduced IL-18 levels and do not develop enhanced atherosclerosis. PLoS One 2010; 5:e13539. [PMID: 21042583 PMCID: PMC2962638 DOI: 10.1371/journal.pone.0013539] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2010] [Accepted: 09/23/2010] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Atherosclerosis is a chronic inflammatory disease that evolves from the interaction of activated endothelial cells, macrophages, lymphocytes and modified lipoproteins (LDLs). In the last years many molecules with crucial metabolic functions have been shown to prevent important steps in the progression of atherogenesis, including peroxisome proliferator activated receptors (PPARs) and the class III histone deacetylase (HDAC) SIRT1. The PPARγ coactivator 1 alpha (Ppargc1a or PGC-1α) was identified as an important transcriptional cofactor of PPARγ and is activated by SIRT1. The aim of this study was to analyze total PGC-1α deficiency in an atherosclerotic mouse model. METHODOLOGY/PRINCIPAL FINDINGS To investigate if total PGC-1α deficiency affects atherosclerosis, we compared ApoE(-/-) PGC-1α(-/-) and ApoE(-/-) PGC-1α(+/+) mice kept on a high cholesterol diet. Despite having more macrophages and a higher ICAM-1 expression in plaques, ApoE(-/-) PGC-1α(-/-) did not display more or larger atherosclerotic plaques than their ApoE(-/-) PGC-1α(+/+) littermates. In line with the previously published phenotype of PGC-1α(-/-) mice, ApoE(-/-) PGC-1α(-/-) mice had marked reduced body, liver and epididymal white adipose tissue (WAT) weight. VLDL/LDL-cholesterol and triglyceride contents were also reduced. Aortic expression of PPARα and PPARγ, two crucial regulators for adipocyte differentiation and glucose and lipid metabolism, as well as the expression of some PPAR target genes was significantly reduced in ApoE(-/-) PGC-1α(-/-) mice. Importantly, the epididymal WAT and aortic expression of IL-18 and IL-18 plasma levels, a pro-atherosclerotic cytokine, was markedly reduced in ApoE(-/-) PGC-1α(-/-) mice. CONCLUSIONS/SIGNIFICANCE ApoE(-/-) PGC-1α(-/-) mice, similar as PGC-1α(-/-) mice exhibit markedly reduced total body and visceral fat weight. Since inflammation of visceral fat is a crucial trigger of atherogenesis, decreased visceral fat in PGC-1α-deficient mice may explain why these mice do not develop enhanced atherosclerosis.
Collapse
Affiliation(s)
- Sokrates Stein
- Cardiovascular Research, Institute of Physiology, and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich and Cardiology, Cardiovascular Center, University Hospital Zurich, Zurich, Switzerland
| | - Christine Lohmann
- Cardiovascular Research, Institute of Physiology, and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich and Cardiology, Cardiovascular Center, University Hospital Zurich, Zurich, Switzerland
| | | | - Elin Stenfeldt
- Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Goteborg, Goteborg, Sweden
| | - Jan Borén
- Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Goteborg, Goteborg, Sweden
| | - Thomas F. Lüscher
- Cardiovascular Research, Institute of Physiology, and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich and Cardiology, Cardiovascular Center, University Hospital Zurich, Zurich, Switzerland
| | - Christian M. Matter
- Cardiovascular Research, Institute of Physiology, and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich and Cardiology, Cardiovascular Center, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
41
|
Hartford M, Wiklund O, Hultén LM, Persson A, Karlsson T, Herlitz J, Hulthe J, Caidahl K. Interleukin-18 as a Predictor of Future Events in Patients With Acute Coronary Syndromes. Arterioscler Thromb Vasc Biol 2010; 30:2039-46. [DOI: 10.1161/atvbaha.109.202697] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
The aim of this study was to assess the short- and long-term prognostic significance of interleukin-18 (IL-18) levels in patients with acute coronary syndromes (ACS).
Methods and Results—
In patients hospitalized with ACS (median age, 66 years; 30% females), we evaluated associations of serum IL-18 levels from day 1 (n=1261) with the short- (<3 months) and long-term (median, 7.6 years) risk of death, development of congestive heart failure (CHF), and myocardial infarction (MI). IL-18 was not significantly associated with short-term mortality. In the long term, IL-18 levels were significantly related to all-cause mortality, even after adjustment for clinical confounders (hazard ratio [HR], 1.19; 95% confidence interval, 1.07 to 1.33;
P
=0.002). Long-term, cardiovascular mortality was univariately related to IL-18, and the adjusted relation between noncardiovascular mortality and IL-18 was highly significant (HR, 1.36; 95% confidence interval, 1.11 to 1.67;
P
=0.003). IL-18 independently predicted CHF, MI, and cardiovascular death/CHF/MI in both the short and long term. Measurements from day 1 of ACS and 3 months after ACS had a similar power to predict late outcome.
Conclusion—
The addition of the measurement of IL-18 to clinical variables improved the prediction of risk of all-cause and noncardiovascular mortality. The association between IL-18 and noncardiovascular mortality is intriguing and warrants further study.
Collapse
Affiliation(s)
- Marianne Hartford
- From the Institute of Medicine, Department of Molecular and Clinical Medicine (M.H., A.P.T.K., J. Herlitz, K.C.), Wallenberg Laboratory for Cardiovascular Research (O.W., L.M.H., J. Hulthe), and Department of Clinical Physiology (A.P., K.C.), Sahlgrenska Academy and Sahlgrenska University Hospital, Gothenburg, Sweden; AstraZeneca R&D, Mölndal, Sweden (J. Hulthe); and the Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden (K.C.)
| | - Olov Wiklund
- From the Institute of Medicine, Department of Molecular and Clinical Medicine (M.H., A.P.T.K., J. Herlitz, K.C.), Wallenberg Laboratory for Cardiovascular Research (O.W., L.M.H., J. Hulthe), and Department of Clinical Physiology (A.P., K.C.), Sahlgrenska Academy and Sahlgrenska University Hospital, Gothenburg, Sweden; AstraZeneca R&D, Mölndal, Sweden (J. Hulthe); and the Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden (K.C.)
| | - Lillemor Mattsson Hultén
- From the Institute of Medicine, Department of Molecular and Clinical Medicine (M.H., A.P.T.K., J. Herlitz, K.C.), Wallenberg Laboratory for Cardiovascular Research (O.W., L.M.H., J. Hulthe), and Department of Clinical Physiology (A.P., K.C.), Sahlgrenska Academy and Sahlgrenska University Hospital, Gothenburg, Sweden; AstraZeneca R&D, Mölndal, Sweden (J. Hulthe); and the Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden (K.C.)
| | - Anita Persson
- From the Institute of Medicine, Department of Molecular and Clinical Medicine (M.H., A.P.T.K., J. Herlitz, K.C.), Wallenberg Laboratory for Cardiovascular Research (O.W., L.M.H., J. Hulthe), and Department of Clinical Physiology (A.P., K.C.), Sahlgrenska Academy and Sahlgrenska University Hospital, Gothenburg, Sweden; AstraZeneca R&D, Mölndal, Sweden (J. Hulthe); and the Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden (K.C.)
| | - Thomas Karlsson
- From the Institute of Medicine, Department of Molecular and Clinical Medicine (M.H., A.P.T.K., J. Herlitz, K.C.), Wallenberg Laboratory for Cardiovascular Research (O.W., L.M.H., J. Hulthe), and Department of Clinical Physiology (A.P., K.C.), Sahlgrenska Academy and Sahlgrenska University Hospital, Gothenburg, Sweden; AstraZeneca R&D, Mölndal, Sweden (J. Hulthe); and the Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden (K.C.)
| | - Johan Herlitz
- From the Institute of Medicine, Department of Molecular and Clinical Medicine (M.H., A.P.T.K., J. Herlitz, K.C.), Wallenberg Laboratory for Cardiovascular Research (O.W., L.M.H., J. Hulthe), and Department of Clinical Physiology (A.P., K.C.), Sahlgrenska Academy and Sahlgrenska University Hospital, Gothenburg, Sweden; AstraZeneca R&D, Mölndal, Sweden (J. Hulthe); and the Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden (K.C.)
| | - Johannes Hulthe
- From the Institute of Medicine, Department of Molecular and Clinical Medicine (M.H., A.P.T.K., J. Herlitz, K.C.), Wallenberg Laboratory for Cardiovascular Research (O.W., L.M.H., J. Hulthe), and Department of Clinical Physiology (A.P., K.C.), Sahlgrenska Academy and Sahlgrenska University Hospital, Gothenburg, Sweden; AstraZeneca R&D, Mölndal, Sweden (J. Hulthe); and the Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden (K.C.)
| | - Kenneth Caidahl
- From the Institute of Medicine, Department of Molecular and Clinical Medicine (M.H., A.P.T.K., J. Herlitz, K.C.), Wallenberg Laboratory for Cardiovascular Research (O.W., L.M.H., J. Hulthe), and Department of Clinical Physiology (A.P., K.C.), Sahlgrenska Academy and Sahlgrenska University Hospital, Gothenburg, Sweden; AstraZeneca R&D, Mölndal, Sweden (J. Hulthe); and the Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden (K.C.)
| |
Collapse
|
42
|
Reddy VS, Prabhu SD, Mummidi S, Valente AJ, Venkatesan B, Shanmugam P, Delafontaine P, Chandrasekar B. Interleukin-18 induces EMMPRIN expression in primary cardiomyocytes via JNK/Sp1 signaling and MMP-9 in part via EMMPRIN and through AP-1 and NF-kappaB activation. Am J Physiol Heart Circ Physiol 2010; 299:H1242-54. [PMID: 20693392 DOI: 10.1152/ajpheart.00451.2010] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IL-18 and the extracellular matrix metalloproteinase (MMP) inducer (EMMPRIN) stimulate the expression of proinflammatory cytokines and MMPs and are elevated in myocardial hypertrophy, remodeling, and failure. Here, we report several novel findings in primary cardiomyocytes treated with IL-18. First, IL-18 activated multiple transcription factors, including NF-κB (p50 and p65), activator protein (AP)-1 (cFos, cJun, and JunD), GATA, CCAAT/enhancer-binding protein, myocyte-specific enhancer-binding factor, interferon regulatory factor-1, p53, and specific protein (Sp)-1. Second, IL-18 induced EMMPRIN expression via myeloid differentiation primary response gene 88/IL-1 receptor-associated kinase/TNF receptor-associated factor-6/JNK-dependent Sp1 activation. Third, IL-18 induced a number of MMP genes, particularly MMP-9, at a rapid rate as well as tissue inhibitor of metalloproteinase (TIMP)-1 and TIMP-3 at a slower rate. Finally, the IL-18 induction of MMP-9 was mediated in part via EMMPRIN and through JNK- and ERK-dependent AP-1 activation and p38 MAPK-dependent NF-κB activation. These results suggest that the elevated expression of IL-18 during myocardial injury and inflammation may favor EMMPRIN and MMP induction and extracellular matrix degradation. Therefore, targeting IL-18 or its signaling pathways may be of potential therapeutic benefit in adverse remodeling.
Collapse
|
43
|
He M, Cornelis MC, Kraft P, van Dam RM, Sun Q, Laurie CC, Mirel DB, Chasman DI, Ridker PM, Hunter DJ, Hu FB, Qi L. Genome-wide association study identifies variants at the IL18-BCO2 locus associated with interleukin-18 levels. Arterioscler Thromb Vasc Biol 2010; 30:885-90. [PMID: 20150558 PMCID: PMC2841960 DOI: 10.1161/atvbaha.109.199422] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Interleukin-18 (IL-18) is a proinflammatory cytokine involved in the processes of innate and acquired immunities and associated with cardiovascular disease and type 2 diabetes. We sought to identify the common genetic variants associated with IL-18 levels. METHODS AND RESULTS We performed a 2-stage genome-wide association study among women of European ancestry from the Nurses' Health Study (NHS) and Women's Genome Health Study (WGHS). IL-18 levels were measured by ELISA. In the discovery stage (NHS, n=1523), 7 single-nucleotide polymorphisms (SNPs) at the IL18-BCO2 locus were associated with IL-18 concentrations at the 1 x 10(-5) significance level. The strongest association was found for SNP rs2115763 in the BCO2 gene (P=6.31 x 10(-8)). In silico replication in WGHS (435 women) confirmed these findings. The combined analysis of the 2 studies indicated that SNPs rs2115763, rs1834481, and rs7106524 reached a genome-wide significance level (P<5 x 10(-8)). Forward selection analysis indicated that SNPs rs2115763 and rs1834481 were independently associated with IL-18 levels (P=0.0002 and 0.0006, respectively). The 2 SNPs together explained 2.9% of variation of plasma IL-18 levels. CONCLUSIONS This study identified several novel variants at the IL18-BCO2 locus associated with IL-18 levels.
Collapse
Affiliation(s)
- Meian He
- Department of Nutrition, Harvard School of Public Health, Boston 02115, Massachusetts, USA
- Institute of Occupational Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei, China
| | - Marilyn C. Cornelis
- Department of Nutrition, Harvard School of Public Health, Boston 02115, Massachusetts, USA
| | - Peter Kraft
- Program in Molecular and Genetic Epidemiology, Department of Epidemiology, Harvard School of Public Health, Boston 02115, Massachusetts, USA
| | - Rob M. van Dam
- Department of Nutrition, Harvard School of Public Health, Boston 02115, Massachusetts, USA
- Program in Molecular and Genetic Epidemiology, Department of Epidemiology, Harvard School of Public Health, Boston 02115, Massachusetts, USA
- Channing Laboratory, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, 02115, Massachusetts, USA
| | - Qi Sun
- Department of Nutrition, Harvard School of Public Health, Boston 02115, Massachusetts, USA
| | - Cathy C. Laurie
- Department of Biostatistics, University of Washington, Seattle 98195, WA, USA
| | - Daniel B. Mirel
- Center for Genotyping and Analysis, Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge 02142, MA,USA
| | - Daniel I. Chasman
- Center for Cardiovascular Disease Prevention, Brigham and Women’s Hospital, Harvard Medical School, Boston 02115, Massachusetts, USA
- Donald W. Reynolds Center for Cardiovascular Research, Brigham and Women’s Hospital, Harvard Medical School, Boston 02115, Massachusetts, USA
| | - Paul M. Ridker
- Center for Cardiovascular Disease Prevention, Brigham and Women’s Hospital, Harvard Medical School, Boston 02115, Massachusetts, USA
- Donald W. Reynolds Center for Cardiovascular Research, Brigham and Women’s Hospital, Harvard Medical School, Boston 02115, Massachusetts, USA
| | - David J. Hunter
- Program in Molecular and Genetic Epidemiology, Department of Epidemiology, Harvard School of Public Health, Boston 02115, Massachusetts, USA
- Channing Laboratory, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, 02115, Massachusetts, USA
| | - Frank B. Hu
- Department of Nutrition, Harvard School of Public Health, Boston 02115, Massachusetts, USA
- Program in Molecular and Genetic Epidemiology, Department of Epidemiology, Harvard School of Public Health, Boston 02115, Massachusetts, USA
- Channing Laboratory, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, 02115, Massachusetts, USA
| | - Lu Qi
- Department of Nutrition, Harvard School of Public Health, Boston 02115, Massachusetts, USA
- Channing Laboratory, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, 02115, Massachusetts, USA
| |
Collapse
|
44
|
Hernesniemi JA, Heikkilä A, Raitakari OT, Kähönen M, Juonala M, Hutri-Kähönen N, Marniemi J, Viikari J, Lehtimäki T. Interleukin-18 gene polymorphism and markers of subclinical atherosclerosis. The Cardiovascular Risk in Young Finns Study. Ann Med 2010; 42:223-30. [PMID: 20350254 DOI: 10.3109/07853891003769940] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND AND AIM Interleukin-18 (IL-18) is a pro-atherosclerotic cytokine. We wanted to evaluate whether IL-18 gene polymorphism associates independently of risk factors, with early subclinical markers of atherosclerosis (intima-media thickness (IMT), coronary artery compliance (CAC), and flow-mediated dilatation (FMD)) in a population of young healthy Caucasian adults. METHODS This study was based on the on-going Cardiovascular Risk in Young Finns Study consisting of 2260 young adults, mean age being 31.7 (range 24-39 years) (1247 women and 1013 men). RESULTS Five studied tagSNPs formed six major haplotypes, which accounted for 99.9% of all variation of the IL-18 gene. According to adjusted analysis of variance, the IL-18 gene polymorphism did not associate with subclinical atherosclerosis in the whole study population. However, one major haplotype associated differently among men and women with IMT (P = 0.011). Male carriers of a major CCTgT haplotype (n = 441) seemed to have a lower IMT when compared to the non-carriers (-0.016 mm, 95% confidence interval (CI) -0.028 to -0.004, P = 0.014). Among women no significant associations were observed. CONCLUSIONS Among all study subjects, the polymorphism of the IL-18 gene is not associated with subclinical markers of atherosclerosis. However, among men one major IL-18 haplotype seemed to associate with substantially lower IMT values.
Collapse
Affiliation(s)
- Jussi A Hernesniemi
- Department of Clinical Chemistry, University of Tampere and Tampere University Hospital, Finland.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Ferraro S, Marano G, Bongo AS, Boracchi P, Biganzoli EM. From multimarker approach to multiplex assays in acute coronary syndromes: What are we searching for? ACTA ACUST UNITED AC 2010; 12:18-24. [DOI: 10.3109/17482940903578980] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
46
|
Hivert MF, Sun Q, Shrader P, Mantzoros CS, Meigs JB, Hu FB. Circulating IL-18 and the risk of type 2 diabetes in women. Diabetologia 2009; 52:2101-8. [PMID: 19669125 PMCID: PMC3758765 DOI: 10.1007/s00125-009-1455-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Accepted: 06/19/2009] [Indexed: 10/20/2022]
Abstract
AIMS/HYPOTHESIS The pathophysiology of type 2 diabetes involves pro-inflammatory pathways. We tested the hypothesis that IL-18 predicts future diabetes cases. METHODS We used a nested case-control design based in the Nurses' Health Study. Baseline blood samples were collected between 1989 and 1990. Questionnaires to assess body weight, lifestyle (physical activity, diet, smoking) and diabetes diagnosis were sent out and assessed biennially (follow-up until 2002). Cases (n = 1,012) were defined as women developing type 2 diabetes at least 1 year after blood sampling. Control women (n = 1,081) were matched to cases by age, date of blood draw, fasting status and race. We calculated the RR (95% CI) of type 2 diabetes in quintiles of IL-18 using conditional logistic regression with the first quintile as referent; adjustments included matching factors, diabetes risk factors, BMI, adipokine levels (adiponectin, resistin) and inflammatory proteins (C-reactive protein, tumour necrosis factor receptor 2 (TNFalpha-R2) and IL-6). RESULTS Higher IL-18 levels were associated with increased risk of developing diabetes, even after adjustment for matching factors and multiple diabetes risk factors: being in the highest quintile of IL-18 was associated with a RR of 1.75 (1.41-2.18) for diabetes relative to the first quintile (p < 0.0001 for trend). Significant trends in association were still observed after adjustment for BMI (RR 1.44 [1.15-1.80], p < 0.0001 for trend) and adiponectin levels (RR 1.28 [1.02-1.60], p = 0.006 for trend). Further adjustment for inflammatory markers in a sub-sample did not significantly change the results. CONCLUSIONS/INTERPRETATION Elevated IL-18 levels are associated with higher risk of diabetes. This association is independent of usual risk factors, including BMI and adipokine levels.
Collapse
Affiliation(s)
- M F Hivert
- Massachusetts General Hospital, Boston, USA
| | | | | | | | | | | |
Collapse
|
47
|
Welsh P, Woodward M, Rumley A, Lowe G. Associations of circulating TNFα and IL-18 with myocardial infarction and cardiovascular risk markers: The Glasgow Myocardial Infarction Study. Cytokine 2009; 47:143-7. [DOI: 10.1016/j.cyto.2009.06.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2009] [Revised: 06/02/2009] [Accepted: 06/09/2009] [Indexed: 11/27/2022]
|
48
|
Yearley JH, Xia D, Pearson C, Carville A, Shannon RP, Mansfield KG. Interleukin-18 predicts atherosclerosis progression in SIV-infected and uninfected rhesus monkeys (Macaca mulatta) on a high-fat/high-cholesterol diet. J Transl Med 2009; 89:657-67. [PMID: 19381133 PMCID: PMC2764242 DOI: 10.1038/labinvest.2009.29] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Interleukin (IL)-18 levels have been identified as important predictors of cardiovascular mortality and are often elevated in human immunodeficiency virus (HIV)-infected individuals. To investigate a possible function for IL-18 in atherogenesis in the context of early HIV infection, we used the simian immunodeficiency model of HIV infection. Acutely simian immunodeficiency virus-infected and uninfected rhesus monkeys (Macaca mulatta) on an atherogenic diet were evaluated prospectively for atherosclerotic lesion development relative to a panel of plasma markers including IL-18, IL-8, IL-1beta, IL-6, C-reactive protein, soluble vascular cell adhesion molecule-1, soluble E-selectin, and soluble intercellular adhesion molecule-1. Although no significant differences in lesion development were identified between groups after 35 days of infection, levels of plasma IL-18 measured 1 month before virus inoculation correlated significantly with atherosclerotic plaque cross-sectional area at the carotid bifurcation (P<0.001, R=0.946), common iliac bifurcation (P<0.01, R=0.789), and cranial abdominal aorta (P<0.01, R=0.747), as well as with extent of CD3+ and CD68+ cellular infiltration in vascular lesions (both P<0.001, R>or=0.835) in both groups. Atherosclerotic plaque area at the carotid and common iliac bifurcations also showed a weaker inverse correlation with baseline IL-8 levels, as did CD68+ signal area. Results implicate a strong role for IL-18 in early atherosclerosis progression and raise the possibility that the chronically elevated IL-18 levels seen in later stages of HIV infection may contribute significantly to accelerated atherogenesis in this population.
Collapse
Affiliation(s)
- Jennifer H. Yearley
- Harvard Medical School, New England Primate Research Center, Southborough, Massachusetts
| | - Dongling Xia
- Harvard Medical School, New England Primate Research Center, Southborough, Massachusetts
| | - Christine Pearson
- Harvard Medical School, New England Primate Research Center, Southborough, Massachusetts
| | - Angela Carville
- Harvard Medical School, New England Primate Research Center, Southborough, Massachusetts
| | - Richard P. Shannon
- Allegheny General Hospital, Pittsburgh, PA,University of Pennsylvania School of Medicine, Department of Medicine, Philadelphia, PA
| | - Keith G. Mansfield
- Harvard Medical School, New England Primate Research Center, Southborough, Massachusetts
| |
Collapse
|
49
|
The role of interleukin 18 in the pathogenesis of hypertension-induced vascular disease. Nat Rev Cardiol 2009; 6:192-9. [DOI: 10.1038/ncpcardio1453] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2008] [Accepted: 01/07/2009] [Indexed: 02/07/2023]
|
50
|
Samnegård A, Hulthe J, Silveira A, Ericsson CG, Hamsten A, Eriksson P. Gender specific associations between matrix metalloproteinases and inflammatory markers in post myocardial infarction patients. Atherosclerosis 2009; 202:550-6. [DOI: 10.1016/j.atherosclerosis.2008.05.050] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2007] [Revised: 04/28/2008] [Accepted: 05/20/2008] [Indexed: 11/25/2022]
|