1
|
Maaniitty E, Jalkanen J, Sinisilta S, Gunn J, Vasankari T, Biancari F, Jalkanen S, Airaksinen KEJ, Hollmen M, Kiviniemi T. Differential circulating cytokine profiles in acute coronary syndrome versus stable coronary artery disease. Sci Rep 2024; 14:17269. [PMID: 39068298 PMCID: PMC11283453 DOI: 10.1038/s41598-024-68333-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024] Open
Abstract
Chronic inflammation plays a crucial role in coronary artery disease (CAD), but differences in specific cytokine profiles between acute coronary syndrome (ACS) and stable CAD remain unknown. We investigated cytokine differences between these two manifestations of CAD. The study included 308 patients with angiographically detected, hemodynamically significant CAD: 150 patients undergone angiography for ACS, 158 patients undergone angiography for stable CAD. To assess dynamic changes, 116 patients had index angiogram at least 3 months earlier. We measured the serum concentrations of 48 circulating cytokines. The ACS group had decreased interleukin (IL) 4 (p = 0.005), and increased IL-8 (p = 0.008), hepatocyte growth factor (HGF) (p < 0.001) and macrophage colony-stimulating factor (M-CSF) (p = 0.002) levels compared with the stable CAD group. Multivariable logistic regression revealed increased levels of HGF (OR 18.050 [95% CI 4.372-74.517], p < 0.001), M-CSF (OR 2.257 [1.375-3.705], p = 0.001) and IL-6 (OR 1.586 [1.131-2.224], p = 0.007), independently associated with ACS. In the post-angiography group, only diminished platelet-derived growth factor-BB levels in ACS-manifested patients were observed (OR 0.478, [0.279-0.818], p = 0.007). Cytokine profiles differ between ACS and stable CAD. Such differences seem to be mainly reversible within 3 months after ACS. Thus, targeting one or two cytokines only might not offer one-size fits all-therapeutic approach for CAD-associated inflammation.Trial registration: NCT03444259.
Collapse
Affiliation(s)
- Eveliina Maaniitty
- Heart Center, Turku University Hospital and University of Turku, POB 52, 20521, Turku, Finland.
| | - Juho Jalkanen
- Vascular Surgery, Turku University Hospital and University of Turku, POB 52, 20521, Turku, Finland
| | - Sami Sinisilta
- Heart Center, Turku University Hospital and University of Turku, POB 52, 20521, Turku, Finland
| | - Jarmo Gunn
- Heart Center, Turku University Hospital and University of Turku, POB 52, 20521, Turku, Finland
| | - Tuija Vasankari
- Heart Center, Turku University Hospital and University of Turku, POB 52, 20521, Turku, Finland
| | - Fausto Biancari
- Heart Center, Turku University Hospital and University of Turku, POB 52, 20521, Turku, Finland
- Department of Medicine, South Karelia Central Hospital, University of Helsinki, Valto Käkelän Katu 1, 53130, Lappeenranta, Finland
| | - Sirpa Jalkanen
- Medicity Research Laboratory, University of Turku, Tykistökatu 6A, 20520, Turku, Finland
| | - K E Juhani Airaksinen
- Heart Center, Turku University Hospital and University of Turku, POB 52, 20521, Turku, Finland
| | - Maija Hollmen
- Medicity Research Laboratory, University of Turku, Tykistökatu 6A, 20520, Turku, Finland
| | - Tuomas Kiviniemi
- Heart Center, Turku University Hospital and University of Turku, POB 52, 20521, Turku, Finland.
| |
Collapse
|
2
|
Wang C, Wu Y, Su Y, Mao B, Luo Y, Yan Y, Hu K, Lu Y, Che W, Wan M. Elevated levels of sIL-2R, TNF-α and hs-CRP are independent risk factors for post percutaneous coronary intervention coronary slow flow in patients with non-ST segment elevation acute coronary syndrome. Int J Cardiovasc Imaging 2022; 38:1191-1202. [PMID: 35182256 PMCID: PMC11143008 DOI: 10.1007/s10554-022-02529-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 01/12/2022] [Indexed: 11/27/2022]
Abstract
To evaluate the association between circulating levels of inflammatory cytokines and the occurrence of post-percutaneous coronary intervention (PCI) coronary slow flow (CSF) in patients with non-ST segment elevation acute coronary syndrome (NSTE-ACS). CSF after PCI commonly occurs and implies poor outcomes, while the determinants of post-PCI CSF in patients with NSTE-ACS remain controversial. In this multicenter case control study, 176 patients diagnosed with NSTE-ACS and with post-PCI CSF occurred composed of CSF group, while 352 matched NSTE-ACS patients composed control group. Corrected thrombolysis in myocardial infarction frame count (cTFC), circulating levels of inflammatory cytokines and PCI related parameters were analyzed using Logistic regression models. Among 528 patients with median age of 67 (59-76) and male proportion of 65.5%, 176 (35.0%) patients had occurrence of post-PCI CSF defined as cTFC ≥ 24. Patients with CSF presented more intense inflammatory activity revealed by higher levels of white blood cell, high-sensitivity C-reactive protein (hs-CRP), interleukin-1ß (IL-1ß), soluble IL-2 receptor (sIL-2R), IL-6, IL-8, IL-10 and tumor necrosis factor-α (TNF-α), while PCI related parameters were comparable. Correlation analysis showed cTFC was positively correlated with those inflammatory cytokines. Logistic regression model indicates that hs-CRP (odds ratio (OR) = 3.038, 95% confidence interval (CI) 1.545-5.975), sIL-2R (OR = 2.103, 95% CI 1.959-4.026) and TNF-α (OR = 3.708, 95% CI 1.426-9.641) were valuable predictors for CSF occurrence. Elevated circulating levels of inflammatory cytokine including hs-CRP, sIL-2R and TNF-α rather than PCI related parameters could predict post-PCI CSF in patients with NSTE-ACS.
Collapse
Affiliation(s)
- Cheng Wang
- Department of Cardiology, Chongming Branch, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 66 East Xiangyang Road, Chongming, Shanghai, China
| | - Yan Wu
- Department of Cardiology, Shanghai Putuo District Liqun Hospital, Shanghai, 200333, China
| | - Yang Su
- Department of Cardiology, Chongming Branch, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 66 East Xiangyang Road, Chongming, Shanghai, China
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai, 200072, China
| | - Bin Mao
- Department of Cardiology, Chongming Branch, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 66 East Xiangyang Road, Chongming, Shanghai, China
| | - Yihong Luo
- Department of Cardiology, Chongming Branch, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 66 East Xiangyang Road, Chongming, Shanghai, China
| | - Yexiang Yan
- Department of Cardiology, Chongming Branch, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 66 East Xiangyang Road, Chongming, Shanghai, China
| | - Kun Hu
- Department of Cardiology, Chongming Branch, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 66 East Xiangyang Road, Chongming, Shanghai, China
| | - Yi Lu
- Department of Cardiology, Chongming Branch, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 66 East Xiangyang Road, Chongming, Shanghai, China
| | - Wenliang Che
- Department of Cardiology, Chongming Branch, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 66 East Xiangyang Road, Chongming, Shanghai, China.
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai, 200072, China.
| | - Minying Wan
- Department of Cardiology, Chongming Branch, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 66 East Xiangyang Road, Chongming, Shanghai, China.
| |
Collapse
|
3
|
Córdova-Martínez A, Caballero-García A, Pérez-Valdecantos D, Roche E, Noriega-González DC. Peripheral Neuropathies Derived from COVID-19: New Perspectives for Treatment. Biomedicines 2022; 10:1051. [PMID: 35625788 PMCID: PMC9138404 DOI: 10.3390/biomedicines10051051] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/26/2022] [Accepted: 04/30/2022] [Indexed: 12/15/2022] Open
Abstract
Peripheral neuropathies constitute a group of disorders affecting the peripheral nervous system. Neuropathies have multiple causes such as infections (i.e., COVID-19), diabetes, and nutritional (low vitamin levels), among others. Many micronutrients, such as vitamins (A, C, D, E, B6, B12, and folate), certain minerals (Fe, Mg, Zn, Se, and Cu), and ω-3 fatty acids have immunomodulatory effects. Therefore, they may play an instrumental role in the treatment of COVID-19 infection. However, many COVID-19 patients can undergo neuropathy. In this context, there is a wealth of information on a variety of first-, second-, and third-line treatment options. This review focuses on the application of nutraceutical strategies in order to improve the symptomatology of neuropathy and neuropathic pain in patients that suffered from COVID-19. Our aim is to provide an alternative vision to traditional medical-pharmacological treatment through nutraceuticals.
Collapse
Affiliation(s)
- Alfredo Córdova-Martínez
- Department of Biochemistry, Molecular Biology and Physiology, Faculty of Health Sciences, GIR Physical Exercise and Aging, University of Valladolid, Campus Duques de Soria, 42004 Soria, Spain;
| | - Alberto Caballero-García
- Department of Anatomy and Radiology, Faculty of Health Sciences, GIR Physical Exercise and Aging, University of Valladolid, Campus Duques de Soria, 42004 Soria, Spain;
| | - Daniel Pérez-Valdecantos
- Department of Biochemistry, Molecular Biology and Physiology, Faculty of Health Sciences, GIR Physical Exercise and Aging, University of Valladolid, Campus Duques de Soria, 42004 Soria, Spain;
| | - Enrique Roche
- Department of Applied Biology-Nutrition, Institute of Bioengineering, University Miguel Hernández, 03202 Elche, Spain;
- Alicante Institute for Health and Biomedical Research (ISABIAL), 03010 Alicante, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - David César Noriega-González
- Department of Surgery, Ophthalmology, Otorhinolaryngology and Physiotherapy, Faculty of Medicine, Hospital Clínico Universitario de Valladolid, 47005 Valladolid, Spain;
| |
Collapse
|
4
|
Afrin H, Salazar CJ, Kazi M, Ahamad SR, Alharbi M, Nurunnabi M. Methods of screening, monitoring and management of cardiac toxicity induced by chemotherapeutics. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.01.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
5
|
Ling Y, Weng H, Tang S. The relationship between IL-6 levels and the angiographic severity of coronary artery disease following percutaneous coronary intervention in acute coronary syndrome patients. BMC Cardiovasc Disord 2021; 21:578. [PMID: 34861824 PMCID: PMC8642871 DOI: 10.1186/s12872-021-02406-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 11/26/2021] [Indexed: 12/27/2022] Open
Abstract
Background The present investigation was developed for the exploration of the association between IL-6 levels and acute coronary syndrome (ACS) findings upon angiographic evaluation. Methods A retrospective review of 346 patients suffering from chest discomfort that underwent coronary angiography was performed. The SYNergy between Percutaneous Coronary Intervention with TAXus and cardiac surgery (SYNTAX) score (SS) and SS II were used to gauge ACS severity, with ACS patients being stratified into two groups based on an SS value of 22 and the median SS II value. Associations between IL-6 levels and SS or SS II values were assessed through Spearman's correlation analyses, and independent predictors of intermediate-high SS or high SS II were identified via a multivariate logistic regression approach. A receiver operating characteristic (ROC) curve was employed to explore of the predictive value of IL-6 levels. Results IL-6 was positively correlated with both SS (r = 0.479, P < 0.001) and SS II (r = 0.305, P < 0.001). Moreover, IL-6 levels were independently predictive of intermediate-high SS and high SS II values. ROC curves further demonstrated that IL-6 was able to predict intermediate-high SS and high SS II, with area under the curve (AUC) values of 0.806 and 0.624, respectively. Conclusion IL-6 levels are closely linked to the extent of coronary artery disease in ACS patients undergoing percutaneous coronary intervention. IL-6 levels may thus serve as a valuable and non-invasive biomarker of high-risk ACS patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-021-02406-7.
Collapse
Affiliation(s)
- Yang Ling
- Department of Cardiology, Yijishan Hospital Affiliated To Wannan Medical College, 2# West Zhe Shan Road, Wuhu, 241000, China
| | - Hairong Weng
- Department of Emergency Intensive Care Unit, Yijishan Hospital Affiliated To Wannan Medical College, 2# West Zhe Shan Road, Wuhu, 241000, China
| | - Shengxing Tang
- Department of Cardiology, Yijishan Hospital Affiliated To Wannan Medical College, 2# West Zhe Shan Road, Wuhu, 241000, China.
| |
Collapse
|
6
|
Wadhawan A, Reynolds MA, Makkar H, Scott AJ, Potocki E, Hoisington AJ, Brenner LA, Dagdag A, Lowry CA, Dwivedi Y, Postolache TT. Periodontal Pathogens and Neuropsychiatric Health. Curr Top Med Chem 2021; 20:1353-1397. [PMID: 31924157 DOI: 10.2174/1568026620666200110161105] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 12/04/2019] [Accepted: 12/04/2019] [Indexed: 02/08/2023]
Abstract
Increasing evidence incriminates low-grade inflammation in cardiovascular, metabolic diseases, and neuropsychiatric clinical conditions, all important causes of morbidity and mortality. One of the upstream and modifiable precipitants and perpetrators of inflammation is chronic periodontitis, a polymicrobial infection with Porphyromonas gingivalis (P. gingivalis) playing a central role in the disease pathogenesis. We review the association between P. gingivalis and cardiovascular, metabolic, and neuropsychiatric illness, and the molecular mechanisms potentially implicated in immune upregulation as well as downregulation induced by the pathogen. In addition to inflammation, translocation of the pathogens to the coronary and peripheral arteries, including brain vasculature, and gut and liver vasculature has important pathophysiological consequences. Distant effects via translocation rely on virulence factors of P. gingivalis such as gingipains, on its synergistic interactions with other pathogens, and on its capability to manipulate the immune system via several mechanisms, including its capacity to induce production of immune-downregulating micro-RNAs. Possible targets for intervention and drug development to manage distal consequences of infection with P. gingivalis are also reviewed.
Collapse
Affiliation(s)
- Abhishek Wadhawan
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, United States.,Department of Psychiatry, Saint Elizabeths Hospital, Washington, D.C. 20032, United States
| | - Mark A Reynolds
- Department of Advanced Oral Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore 21201, United States
| | - Hina Makkar
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, United States
| | - Alison J Scott
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, United States
| | - Eileen Potocki
- VA Maryland Healthcare System, Baltimore VA Medical Center, Baltimore, United States
| | - Andrew J Hoisington
- Air Force Institute of Technology, Wright-Patterson Air Force Base, United States
| | - Lisa A Brenner
- Departments of Psychiatry, Neurology, and Physical Medicine & Rehabilitation, University of Colorado Anschutz Medical Campus, Aurora, United States.,Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Aurora, United States.,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, United States
| | - Aline Dagdag
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, United States
| | - Christopher A Lowry
- Departments of Psychiatry, Neurology, and Physical Medicine & Rehabilitation, University of Colorado Anschutz Medical Campus, Aurora, United States.,Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Aurora, United States.,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, United States.,Department of Integrative Physiology, Center for Neuroscience and Center for Microbial Exploration, University of Colorado Boulder, Boulder, United States.,Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, United States
| | - Yogesh Dwivedi
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Alabama, United States
| | - Teodor T Postolache
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, United States.,Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Aurora, United States.,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, United States.,Mental Illness Research, Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 5, VA Capitol Health Care Network, Baltimore, United States
| |
Collapse
|
7
|
Salari N, Mansouri K, Hosseinian-Far A, Ghasemi H, Mohammadi M, Jalali R, Vaisi-Raygani A. The effect of polymorphisms (174G> C and 572C> G) on the Interleukin-6 gene in coronary artery disease: a systematic review and meta-analysis. Genes Environ 2021; 43:1. [PMID: 33436103 PMCID: PMC7802194 DOI: 10.1186/s41021-021-00172-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/02/2021] [Indexed: 02/08/2023] Open
Abstract
Background Coronary Artery Disease (CAD) is caused by the blockage of the coronary arteries. it is argued that there has an association between the Interleukin-6 gene and the occurrence of atherosclerosis, coronary artery disease, Due to the short half-life and high variability of Interleukin-6 (IL-6), limited studies have been performed on the association of serum levels of interleukin-6 with coronary artery disease. The aim of this study is to investigate the relationship between IL-6 gene polymorphisms and coronary artery disease. Methods This study was conducted as a meta-analysis of selected articles with no lower time limit and upto March 2020. Articles related to the subject were obtained by searching several data sources,such as the SID, IranDoc, Scopus, Embase, Web of Science (ISI), PubMed, Science Direct, and Google Scholar databases. The heterogeneity of the studies was assessed using the I2 index in the Comprehensive Meta-Analysis software. Results The GG genotype of the IL-6174 G> C polymorphism with a 0.8 odds ratio tended to reduce the risk of CAD by 20%. The odds ratio of CAD in CG and GG genotypes were found to be 1.16 and 1.48 times respectively, indicating the increasing effect of these two genotypes. In the IL-6-572 C>G polymorphism, CG and GG genotypes increased the risk of CAD by 1.21 and 1.27 times respectively, and the CC genotype tended to reduce the risk of CAD by 15%, considering the odds ratio of 0.85. Conclusion This study showed a relationship between IL-6174G> C and Interleukin-6 (IL-6) 572 C>G genes and coronary artery disease. Moreover, the protective effects of GG genotype in IL-6 gene 174 G> C and CC genotype in IL-6 gene 572 C>G gene were reported. The study also confirmed that the CG and CC genotypes of the G>C IL-6174 gene have an increasing effect on coronary artery disease. Moreover, CG and GG genotypes in the IL-6 gene 572 C>G increased the risk of developing CAD. It should be noted that the increased risk of developing CAD was limited to meta-analytic studies in reported literatures.
Collapse
Affiliation(s)
- Nader Salari
- Department of Biostatistics, School of Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kamran Mansouri
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amin Hosseinian-Far
- Department of Business Systems & Operations, University of Northampton, Northampton, UK
| | - Hooman Ghasemi
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Masoud Mohammadi
- Department of Nursing, School of Nursing and Midwifery, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Rostam Jalali
- Department of Nursing, School of Nursing and Midwifery, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Aliakbar Vaisi-Raygani
- Department of Nursing, School of Nursing and Midwifery, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
8
|
Possible Correlations between Atherosclerosis, Acute Coronary Syndromes and COVID-19. J Clin Med 2020; 9:jcm9113746. [PMID: 33233333 PMCID: PMC7700642 DOI: 10.3390/jcm9113746] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 12/28/2022] Open
Abstract
An outbreak of SARS-CoV-2 infection in December 2019 became a major global concern in 2020. Since then, several articles analyzing the course, complications and mechanisms of the infection have appeared. However, there are very few papers explaining the possible correlations between COVID-19, atherosclerosis and acute coronary syndromes. We performed an analysis of PubMed, Cochrane, Google Scholar, and MEDLINE databases. As of September 15, 2020, the results were as follows: for "COVID-19" and "cardiovascular system" we obtained 687 results; for "COVID-19" and "myocardial infarction" together with "COVID-19" and "acute coronary syndrome" we obtained 328 results; for "COVID-19" and "atherosclerosis" we obtained 57 results. Some of them did not fulfill the search criteria or concerned the field of neurology. Only articles written in English, German and Polish were analyzed for a total number of 432 papers. While the link between inflammatory response, COVID- 19 and atherosclerosis still remains unclear, there is evidence that suggests a more likely correlation between them. Practitioners' efforts should be focused on the prevention of excessive inflammatory response and possible complications, while there are limited specific therapeutic options against SARS-CoV-2. Furthermore, special attention should be paid to cardioprotection during the pandemic.
Collapse
|
9
|
Gager GM, Biesinger B, Hofer F, Winter MP, Hengstenberg C, Jilma B, Eyileten C, Postula M, Lang IM, Siller-Matula JM. Interleukin-6 level is a powerful predictor of long-term cardiovascular mortality in patients with acute coronary syndrome. Vascul Pharmacol 2020; 135:106806. [PMID: 33035661 DOI: 10.1016/j.vph.2020.106806] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/23/2020] [Accepted: 10/03/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND The interleukin-6 (IL-6) pathway has a crucial role in the pathogenesis of atherosclerosis, the main cause of cardiovascular diseases. We aimed to characterize the predictive value of inflammatory biomarkers on long-term cardiovascular mortality in patients with acute coronary syndrome (ACS) undergoing percutaneous coronary intervention (PCI). METHODS This prospective observational study included 322 consecutive patients with ACS undergoing PCI. Blood-derived biomarkers IL-6 and high-sensitivity C-reactive protein (hsCRP) were assessed at the time point of ACS. Patients were followed-up for 6 years. Long-term cardiovascular mortality was our primary endpoint. Adjusted Cox-regression analysis was used for prediction of events. RESULTS Elevated IL-6 values (≥3.3 pg/mL) emerged as an independent and the most powerful predictor for cardiovascular mortality: the ROC analysis showed that IL-6 was more accurate for cardiovascular mortality prediction as compared to hsCRP (IL-6: AUC = 0.72; 95%CI: 0.62-0.81; p = 0.009 vs hsCRP: AUC = 0.56; 95%CI: 0.41-0.72; p = 0.445). The positive predictive value of IL-6 for mortality was 9%, the negative predictive value 99%, sensitivity 94% and specificity 48%. The primary endpoint of long-term cardiovascular death occurred more frequently in patients with high vs low IL-6 (9.0% vs 0.5%, p = 0.001). The multivariate Cox regression analysis revealed that patients with high IL-6 (≥3.3 pg/mL) values were at 8.6-fold higher hazard to die than those with low IL-6 (<3.3 pg/mL) levels (adj. hazard ratio [HR] = 8.60, 95%CI: 1.07-69.32; p = 0.043). CONCLUSION In the setting of ACS, high IL-6 values are associated with substantial long-term cardiovascular mortality. Further, IL-6 performs as a superior predictor for cardiovascular death as compared to hsCRP.
Collapse
Affiliation(s)
- Gloria M Gager
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria; Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Benedikt Biesinger
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Felix Hofer
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Max-Paul Winter
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Christian Hengstenberg
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Ceren Eyileten
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CEPT), Medical University of Warsaw, Warsaw, Poland
| | - Marek Postula
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CEPT), Medical University of Warsaw, Warsaw, Poland
| | - Irene M Lang
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Jolanta M Siller-Matula
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria; Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology (CEPT), Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
10
|
Li H, Cen K, Sun W, Feng B. Predictive Value of Blood Interleukin-6 Level in Patients with Acute Coronary Syndrome: A Meta-analysis. Immunol Invest 2020; 50:964-976. [PMID: 32811241 DOI: 10.1080/08820139.2020.1795876] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Conflicting results have been reported on the association between blood level of interleukin-6 and adverse outcomes in patients with acute coronary syndrome (ACS). The current meta-analysis aimed to evaluate the predictive utility of elevated blood interleukin-6 level in patients with ACS. METHODS A systematically literature search was performed using PubMed and Embase databases up to December 31, 2019. Observational studies or post hoc analysis of randomized controlled trials investigating the values of blood interleukin-6 level for predicting major adverse cardiovascular events (MACE including death, re-infarction, revascularization, angina, heart failure, malignant arrhythmia, or stroke), all-cause mortality or cardiovascular mortality in ACS patients were eligible. The predictive values were summarized by pooling the multivariable-adjusted risk ratio (RR) and 95% confidence intervals (CI) for the highest versus lowest category of interleukin-6 level. RESULTS Thirteen studies enrolling 30,289 patients with ACS were included. When comparing the highest with lowest category of interleukin-6 level, the pooled RR was 1.29 (95% CI 1.12-1.48) for MACE, 1.50 (95% CI 1.35-1.67) for all-cause mortality, and 1.55 (95% CI 1.06-2.28) for cardiovascular mortality, respectively. Moreover, the predictive values of interleukin-6 level on MACE were consistently found in different study designs, subtypes of patients, sample sizes, follow-up duration, and cutoff value of interleukin-6 elevation subgroups. CONCLUSION Increased blood level of interleukin-6may be independently associated with higher risk of MACE, cardiovascular and all-cause mortality in patients with ACS. Measurement of blood interleukin-6 level has potential to improve risk stratification of ACS.
Collapse
Affiliation(s)
- Hengdong Li
- Department of Cardiology, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, China.,Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
| | - Kaidong Cen
- Department of Cardiology, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Weifeng Sun
- Department of Cardiology, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Beili Feng
- Department of Cardiology, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, China.,Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
| |
Collapse
|
11
|
Hampel H, Caraci F, Cuello AC, Caruso G, Nisticò R, Corbo M, Baldacci F, Toschi N, Garaci F, Chiesa PA, Verdooner SR, Akman-Anderson L, Hernández F, Ávila J, Emanuele E, Valenzuela PL, Lucía A, Watling M, Imbimbo BP, Vergallo A, Lista S. A Path Toward Precision Medicine for Neuroinflammatory Mechanisms in Alzheimer's Disease. Front Immunol 2020; 11:456. [PMID: 32296418 PMCID: PMC7137904 DOI: 10.3389/fimmu.2020.00456] [Citation(s) in RCA: 189] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 02/27/2020] [Indexed: 12/13/2022] Open
Abstract
Neuroinflammation commences decades before Alzheimer's disease (AD) clinical onset and represents one of the earliest pathomechanistic alterations throughout the AD continuum. Large-scale genome-wide association studies point out several genetic variants—TREM2, CD33, PILRA, CR1, MS4A, CLU, ABCA7, EPHA1, and HLA-DRB5-HLA-DRB1—potentially linked to neuroinflammation. Most of these genes are involved in proinflammatory intracellular signaling, cytokines/interleukins/cell turnover, synaptic activity, lipid metabolism, and vesicle trafficking. Proteomic studies indicate that a plethora of interconnected aberrant molecular pathways, set off and perpetuated by TNF-α, TGF-β, IL-1β, and the receptor protein TREM2, are involved in neuroinflammation. Microglia and astrocytes are key cellular drivers and regulators of neuroinflammation. Under physiological conditions, they are important for neurotransmission and synaptic homeostasis. In AD, there is a turning point throughout its pathophysiological evolution where glial cells sustain an overexpressed inflammatory response that synergizes with amyloid-β and tau accumulation, and drives synaptotoxicity and neurodegeneration in a self-reinforcing manner. Despite a strong therapeutic rationale, previous clinical trials investigating compounds with anti-inflammatory properties, including non-steroidal anti-inflammatory drugs (NSAIDs), did not achieve primary efficacy endpoints. It is conceivable that study design issues, including the lack of diagnostic accuracy and biomarkers for target population identification and proof of mechanism, may partially explain the negative outcomes. However, a recent meta-analysis indicates a potential biological effect of NSAIDs. In this regard, candidate fluid biomarkers of neuroinflammation are under analytical/clinical validation, i.e., TREM2, IL-1β, MCP-1, IL-6, TNF-α receptor complexes, TGF-β, and YKL-40. PET radio-ligands are investigated to accomplish in vivo and longitudinal regional exploration of neuroinflammation. Biomarkers tracking different molecular pathways (body fluid matrixes) along with brain neuroinflammatory endophenotypes (neuroimaging markers), can untangle temporal–spatial dynamics between neuroinflammation and other AD pathophysiological mechanisms. Robust biomarker–drug codevelopment pipelines are expected to enrich large-scale clinical trials testing new-generation compounds active, directly or indirectly, on neuroinflammatory targets and displaying putative disease-modifying effects: novel NSAIDs, AL002 (anti-TREM2 antibody), anti-Aβ protofibrils (BAN2401), and AL003 (anti-CD33 antibody). As a next step, taking advantage of breakthrough and multimodal techniques coupled with a systems biology approach is the path to pursue for developing individualized therapeutic strategies targeting neuroinflammation under the framework of precision medicine.
Collapse
Affiliation(s)
- Harald Hampel
- Sorbonne University, GRC no. 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France
| | - Filippo Caraci
- Department of Drug Sciences, University of Catania, Catania, Italy.,Oasi Research Institute-IRCCS, Troina, Italy
| | - A Claudio Cuello
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.,Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada.,Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | | | - Robert Nisticò
- Laboratory of Neuropharmacology, EBRI Rita Levi-Montalcini Foundation, Rome, Italy.,School of Pharmacy, Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Massimo Corbo
- Department of Neurorehabilitation Sciences, Casa Cura Policlinico, Milan, Italy
| | - Filippo Baldacci
- Sorbonne University, GRC no. 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France.,Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital, Paris, France.,Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France.,Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Nicola Toschi
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy.,Department of Radiology, "Athinoula A. Martinos" Center for Biomedical Imaging, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States
| | - Francesco Garaci
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy.,Casa di Cura "San Raffaele Cassino", Cassino, Italy
| | - Patrizia A Chiesa
- Sorbonne University, GRC no. 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France.,Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital, Paris, France.,Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | | | | | - Félix Hernández
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.,Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Jesús Ávila
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.,Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | | | | | - Alejandro Lucía
- Faculty of Sport Sciences, Universidad Europea de Madrid, Madrid, Spain.,Research Institute of the Hospital 12 de Octubre ("imas"), Madrid, Spain.,Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | | | - Bruno P Imbimbo
- Research & Development Department, Chiesi Farmaceutici, Parma, Italy
| | - Andrea Vergallo
- Sorbonne University, GRC no. 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France
| | - Simone Lista
- Sorbonne University, GRC no. 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France.,Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital, Paris, France.,Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| |
Collapse
|
12
|
Li J, Wu J, Zhang M, Zheng Y. Dynamic changes of innate lymphoid cells in acute ST-segment elevation myocardial infarction and its association with clinical outcomes. Sci Rep 2020; 10:5099. [PMID: 32198366 PMCID: PMC7083894 DOI: 10.1038/s41598-020-61903-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 03/03/2020] [Indexed: 12/28/2022] Open
Abstract
An increasing body of evidence has implicated the innate immune system in the causation of acute ST-segment elevation myocardial infarction (STEMI). Innate lymphoid cells (ILCs) are newly identified members of the lymphoid lineage that are important effectors of innate immunity. The role of ILCs in STEMI has not been explored. We characterized the ILCs present in peripheral blood of 176 STEMI patients and 52 controls. Patients were followed up for up to 23 months. Flow cytometry showed that the proportion of total ILCs and ILC1s were significantly increased compared with controls; contrary to ILC1s, the proportion of ILC2s among total ILCs decreased significantly during the acute phase of STEMI. ILC1s percentage was an independent predictor of major adverse cardiovascular events (MACE). On multivariate Cox regression, the 3rd tertile of ILC1s was associated with a higher MACE rate compared with the 1st tertile (hazard ratio: 2.26; 95% confidence interval 1.56–3.27; P = 0.014). RNA-sequencing (RNA-Seq) revealed increased expressions of interferon-γ, tumor necrosis factor-α, vascular cell adhesion molecule 1 (VCAM1), and matrix metallopeptidase 9. Moreover, as active factors secreted by ILC1s, levels of interleukin (IL)−12 and IL-18 were significantly increased in STEMI patients. Increased ILC1s in patients with STEMI was associated with poor outcomes. Our findings suggest that ILC1s may play an important role in STEMI.
Collapse
Affiliation(s)
- Jing Li
- Department of Cardiovascular disease, The First Hospital, Jilin University, Changchun, China
| | - Jing Wu
- Translational Medicine, The First Hospital, Jilin University, Changchun, China
| | - Mingyou Zhang
- Department of Cardiovascular disease, The First Hospital, Jilin University, Changchun, China.
| | - Yang Zheng
- Department of Cardiovascular disease, The First Hospital, Jilin University, Changchun, China.
| |
Collapse
|
13
|
YANG B, ZHENG C, YU H, ZHANG R, LI S, LEN M, CAI S. Comparison of Efficacy between Clopidogrel and Ticagrelor in Patients with Acute Coronary Syndrome after Interventional Treatment and Their Effects on IL-6. IRANIAN JOURNAL OF PUBLIC HEALTH 2020; 49:240-248. [PMID: 32461931 PMCID: PMC7231712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
BACKGROUND We aimed to compare the efficacy between clopidogrel and ticagrelor in patients with acute coronary syndrome (ACS) after percutaneous coronary intervention (PCI) and their effects on IL-6. METHODS A retrospective analysis and collection of 200 ACS patients diagnosed by the Department of Cardiology, Affiliated Hospital of Qingdao University, Qingdao, China in 2016 were performed. Patients were randomly divided into clopidogrel group and ticagrelor group. Data of left ventricular ejection fraction and ACS clinical classification before PCI, PCI treatment, IL-6, platelet aggregation status, maximum platelet aggregation rate (MPAR), P2Y12 response unit (PRU) and adverse reaction of patients were collected. After PCI, patients were followed up for 1 year to compare the ischemia after treatment between clopidogrel group and tigravilol group. RESULTS MPAR and PRU after PCI of clopidogrel group were significantly higher than those of ticagrelor group (P<0.05). The expression of IL-6 in two groups peaked at 1 day after PCI and then decreased. That of ticagrelor group was consistently lower than that of clopidogrel group (P<0.05). The incidence of ischemic events after treatment in clopidogrel group was significantly higher than that in ticagrelor group (P<0.001). CONCLUSION Compared with clopidogrel, tigerrilol had more significant inhibition of platelet aggregation after PCI in ACS patients, and tigerrilol had better effect after interventional treatment in ACS patients. In addition, compared with clopidogrel, tegrel can significantly inhibit the expression of IL-6 in patients with ACS and better alleviate the inflammatory response after PCI.
Collapse
Affiliation(s)
- Bin YANG
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chunyan ZHENG
- Medical Consultation Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Haichu YU
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Rui ZHANG
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shan LI
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Min LEN
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shanglang CAI
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China,Corresponding Author:
| |
Collapse
|
14
|
Gu J, Zhu H, Zhu D, Li M, Xiao M, Yan D, Shen S. VWF, CXCL8 and IL6 might be potential druggable genes for acute coronary syndrome (ACS). Comput Biol Chem 2019; 83:107125. [DOI: 10.1016/j.compbiolchem.2019.107125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 09/02/2019] [Accepted: 09/10/2019] [Indexed: 01/31/2023]
|
15
|
Xu X, Liu X, Yu L, Ma J, Yu S, Ni M. Impact of intracoronary nicorandil before stent deployment in patients with acute coronary syndrome undergoing percutaneous coronary intervention. Exp Ther Med 2019; 19:137-146. [PMID: 31853283 PMCID: PMC6909796 DOI: 10.3892/etm.2019.8219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 11/07/2019] [Indexed: 12/29/2022] Open
Abstract
The present study aimed to clarify the effect of bolus intracoronary nicorandil on inflammatory, oxidative and adherent indicators in patients with acute coronary syndrome (ACS) undergoing percutaneous coronary intervention (PCI). This randomized controlled trial (RCT) was performed to detect the inflammation and oxidative stress in intracoronary blood both before and after PCI. In total, 65 consecutive patients undergoing PCI were classified into a nicorandil therapy group (n=32) or a placebo group (n=33). All procedures were performed at Shandong University Qilu Hospital, China, during the period from March, 2016 to May, 2017. Intracoronary blood from patients who received nicorandil therapy during PCI showed no change in soluble CD40 ligand (sCD40L) concentration (1.86±0.08 vs. 1.90±0.09 ng/ml, P=0.12) but a significant increase was noted in the control group (1.87±0.17 vs. 2.82±0.26 ng/ml, P<0.01). This indicated a relative reduction in sCD40L level after PCI in the nicorandil group. We further demonstrated an increase in superoxide dismutase (SOD) activity (29.37±0.81 vs. 31.03±0.60 U/ml, P<0.001) and a reduction in lipid peroxidation (3.84±0.99 vs. 4.23±0.13 U/ml, P=0.001) in the nicorandil group but observed no change in the placebo group. ICAM-1 levels showed no change in the nicorandil group (69.54±6.89 vs. 72.01±8.25 ng/ml, P=0.83) but a significant increase in the control group after PCI in intracoronary blood (56.57±4.96 vs. 76.81±6.88 ng/ml, P=0.002). No changes were found in hs-CRP, TNFα and sVCAM-1 levels in coronary blood for both groups before and after PCI in ACS patients. Our findings demonstrate that intracoronary bolus nicorandil therapy has a significant effect on the inhibition of inflammatory indicators and oxidative stress in patients with ACS during PCI. This suggests a possible medical application of nicorandil for reducing inflammation and oxidative stress.
Collapse
Affiliation(s)
- Xingli Xu
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xiaoling Liu
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Liwen Yu
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Jing Ma
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Sufang Yu
- Department of Neurology, The Fourth People's Hospital, Liaocheng, Shandong 252002, P.R. China
| | - Mei Ni
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
16
|
Bagavad Gita J, George AV, Pavithra N, Chandrasekaran SC, Latchumanadhas K, Gnanamani A. Dysregulation of miR-146a by periodontal pathogens: A risk for acute coronary syndrome. J Periodontol 2019; 90:756-765. [PMID: 30618100 DOI: 10.1002/jper.18-0466] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 12/06/2018] [Accepted: 12/13/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Periodontitis is a polymicrobial, chronic inflammatory disease leading to loss of tooth-supporting structures. The bacteremia, endotoxemia, and systemic low-grade inflammation associate periodontitis with systemic illnesses such as diabetes mellitus and coronary artery disease. Periodontal pathogens have been detected from atheromatous plaque by amplification of the genetic material by using specific oligonucleotide primers in polymerase chain reaction. Though the association between periodontitis and cardiovascular diseases has been ascertained by systematic reviews and meta-analyses, its pathophysiology is not lucid. MicroRNAs are currently implicated in the regulation of many cellular processes including inflammation and may play a vital role in our understanding of this disease association. In this case-control study, we explored the role of the inflammatory microRNA, miR-146a, in acute coronary syndrome (ACS) subjects with and without chronic periodontitis (CP) and its regulation of the innate immune host response to periodontal pathogens. METHODS Three groups each comprising 66 patients each, namely group 1 (ACS patients without CP), group 2 (ACS patients with CP) and group 3 (CP only) formed the study population. Subgingival plaque samples and serum samples were subjected to quantitative Polymerase Chain Reaction (qPCR) for detection of Porphyromonas gingivalis, a keystone pathogen and to assess the levels of circulating miR-146a and associated proinflammatory cytokines. RESULTS miR-146a associated significantly in group 2 subjects with an odds ratio 1.434, 95% confidence interval 1.013-2.030, P < 0.042, and a predictive percentage of 83.3% and group 1 with a predictive percentage of 76.0.% The associated cytokines interleukin-6 (IL-6), tumor necrosis factor-α, and IL-1β also showed an upregulation with statistical significance (P < 0.05). CONCLUSION microRNA-146a is a key molecule associating periodontitis with acute coronary syndrome.
Collapse
Affiliation(s)
- J Bagavad Gita
- Microbiology Division, Biological Materials Laboratory, CSIR-CLRI, Chennai, India.,Department of Periodontology, Sree Balaji Dental College and Hospital, Chennai, India
| | - Ann V George
- Department of Periodontology, Sree Balaji Dental College and Hospital, Chennai, India
| | - N Pavithra
- Microbiology Division, Biological Materials Laboratory, CSIR-CLRI, Chennai, India
| | - S C Chandrasekaran
- Department of Periodontology, Sree Balaji Dental College and Hospital, Chennai, India
| | - K Latchumanadhas
- Department of Adult Cardiology, Madras Medical Mission, Chennai, India
| | - A Gnanamani
- Microbiology Division, Biological Materials Laboratory, CSIR-CLRI, Chennai, India
| |
Collapse
|
17
|
Kozel M, Kočka V, Lisa L, Buděšínský T, Toušek P. Immune-inflammatory response after bioresorbable vascular scaffold implantation in patients with acute myocardial infarction with ST elevation in a long-term perspective. Heart Vessels 2018; 34:557-563. [PMID: 30315494 DOI: 10.1007/s00380-018-1281-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 10/05/2018] [Indexed: 11/25/2022]
Abstract
A higher rate of bioresorbable vascular scaffold (BVS) thrombosis has been observed after device implantation compared to implantation of permanent metallic stents in recently published studies. The mechanism of BVS thrombosis is currently under debate. To assess whether the immune-inflammatory response after BVS implantation is a potential trigger of BVS thrombosis. The PRAGUE-19 study was an academic study that enrolled consecutive patients with ST-segment elevation myocardial infarction (STEMI) with the intention to implant a BVS. A laboratory sub-study included 49 patients with an implanted BVS (of which 38 underwent the complete 2-year follow-up) and 52 patients having an implanted permanent metallic stent as the control group (of which 30 underwent the complete 2-year follow-up). Samples for inflammatory markers [high-sensitivity C-reactive protein (hs-CRP), interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α)] were taken before BVS or stent implantation, on days 1 and 2 after device implantation and at 1 month and 2 years for a clinical control. The primary combined clinical endpoint of the sub-study (death, reinfarction or target vessel revascularization) occurred in 4.08% of the BVS group and 7.69% of the control group (p = 0.442) during the 2-year follow-up period, with overall mortality of 2.04% in the BVS group and 1.92% in the control group (p = 0.966). Definite BVS thrombosis occurred in one patient in the subacute phase; there was no late or very late thrombosis. Two definite stent thromboses were observed in the control group: one in the subacute phase and the other in the late phase. Baseline inflammatory marker levels did not differ between the groups. Lower levels of IL-6 and hs-CRP were observed in the BVS group compared to the control group (12.02 ± 5.94 vs. 15.21 ± 5.33 pg/ml; p < 0.01; 3952.9 ± 1704.75 ng/ml vs. 4507.49 ± 1190.01 ng/ml; p = 0.037, respectively) on days 1 and 2 (12.01 ± 6.31 vs. 13.85 ± 6.01 pg/ml; p = 0.089; 4447.92 ± 1325.31 ng/ml vs. 4637.03 ± 1290.99 ng/ml; p = 0.255, respectively). No differences in IL-6 or hs-CRP were observed after 1 month or 2 years in the clinical control. Levels of TNF-α did not differ between the groups in the early period after BVS or metallic stent implantation, nor during follow-up. The immune-inflammatory response is lower during the early phase after BVS implantation compared to that after metallic stent implantation, but the responses did not differ in the long term.
Collapse
Affiliation(s)
- Martin Kozel
- Cardiocenter, Third Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Ruská 87, Prague 10, Czech Republic
| | - Viktor Kočka
- Cardiocenter, Third Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Ruská 87, Prague 10, Czech Republic
| | - Libor Lisa
- Cardiocenter, Third Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Ruská 87, Prague 10, Czech Republic
| | - Tomáš Buděšínský
- Cardiocenter, Third Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Ruská 87, Prague 10, Czech Republic
| | - Petr Toušek
- Cardiocenter, Third Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Ruská 87, Prague 10, Czech Republic.
| |
Collapse
|
18
|
Ueland T, Kleveland O, Michelsen AE, Wiseth R, Damås JK, Aukrust P, Gullestad L, Halvorsen B, Yndestad A. Serum PCSK9 is modified by interleukin-6 receptor antagonism in patients with hypercholesterolaemia following non-ST-elevation myocardial infarction. Open Heart 2018; 5:e000765. [PMID: 30258647 PMCID: PMC6150185 DOI: 10.1136/openhrt-2017-000765] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 04/06/2018] [Accepted: 05/15/2018] [Indexed: 12/11/2022] Open
Abstract
Objective It is unclear if activation of inflammatory pathways regulates proprotein convertase subtilisin-kexin type 9 (PCSK9) levels. Approach We evaluated (1) the temporal course of serum PCSK9 during hospitalisation following acute coronary syndrome and associations with markers of inflammation (leucocyte counts, interleukin (IL)-6, C-reactive protein) and lipid levels and (2) the effect of inhibition of IL-6 signalling with the IL-6 receptor antibody tocilizumab on PCSK9 levels in a randomised, double-blind, placebo-controlled trial release in patients with non-ST-elevation myocardial infarction. Results Serum PCSK9 increased during the acute phase and this response was modestly associated with neutrophil counts (r=0.24, p=0.009) and presence of hypercholesterolaemia (r=0.019, p=0.045), but was not modified by tocilizumab. However, a modifying effect of tocilizumab on PCSK9 levels was observed in patients with hypercholesterolaemia (p=0.024, repeated measures analysis of variance) and this effect was strongly correlated with the decrease in neutrophils (r=0.66, p=0.004). Conclusions Our study suggests that patients with a more atherogenic profile may benefit from anti-IL-6 therapy with regard to PCSK9. Trial registration number NCT01491074.
Collapse
Affiliation(s)
- Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,KG Jebsen TREC, University of Tromsø, Tromsø, Norway
| | - Ola Kleveland
- Department of Clinic of Cardiology, St Olavs Hospital, Trondheim, Norway.,Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Annika E Michelsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Rune Wiseth
- Department of Clinic of Cardiology, St Olavs Hospital, Trondheim, Norway.,Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Jan Kristian Damås
- Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Lars Gullestad
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Arne Yndestad
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Center for Heart Failure Research, University of Oslo, Oslo, Norway
| |
Collapse
|
19
|
Mozzini C, Garbin U, Stranieri C, Salandini G, Pesce G, Fratta Pasini AM, Cominacini L. Nuclear factor kappa B in patients with a history of unstable angina: case re-opened. Intern Emerg Med 2018; 13:699-707. [PMID: 29858968 DOI: 10.1007/s11739-018-1885-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 05/26/2018] [Indexed: 02/07/2023]
Abstract
This study aims at assessing NF-kB activity in unstable angina (UA) patients free of symptoms after a 1 year follow-up (1YFU). Plasma oxidized low-density lipoproteins (oxLDL), circulating NF-kB, Interleukin 6 (IL-6) and Interleukin 1β (IL-1β), high-sensitivity C-reactive protein (hs-CRP), as markers of oxidative stress and inflammation and plasma double-stranded DNA (ds-DNA), as marker of Neutrophil Extracellular Traps (NETs), were measured in 23 of the previously enrolled 27 UA patients. These measurements were compared to the UA data at baseline, and then compared to the data derived from the stable angina (SA) and controls (C) enrolled in our previous study (we demonstrated that UA had higher levels of NF-kB compared to SA and C). After a 1YFU, UA patients show a significant decrease in NF-kB, IL-6, hs-CRP, oxLDL, and ds-DNA plasma levels (p < 0.001) and in IL-1β and White Blood Cells (WBC) (p < 0.005), without differences in lipid and glucose assessment. If compared to SA and C, UA after a 1YFU have higher levels of NF-kB, IL-6, ds-DNA, WBC, and oxLDL compared to C (p < 0.001), but only IL-6 is higher than SA (p < 0.001). No differences are found in lipid and glucose assessment. After a 1YFU, patients with a history of UA improve their oxidative and inflammatory status, such as the levels of circulating ds-DNA, without achieving the status of C. They become comparable to SA subjects. This study provides new insight on the multiple and apparently contradictory facets of NF-kB in UA and on its possible role as mediator in NETs' formation.
Collapse
Affiliation(s)
- Chiara Mozzini
- Department of Medicine, Section of Internal Medicine, University of Verona, Piazzale L.A. Scuro, 10 - 37134, Verona, Italy.
| | - Ulisse Garbin
- Department of Medicine, Section of Internal Medicine, University of Verona, Piazzale L.A. Scuro, 10 - 37134, Verona, Italy
| | - Chiara Stranieri
- Department of Medicine, Section of Internal Medicine, University of Verona, Piazzale L.A. Scuro, 10 - 37134, Verona, Italy
| | - Giulia Salandini
- Department of Medicine, Section of Internal Medicine, University of Verona, Piazzale L.A. Scuro, 10 - 37134, Verona, Italy
| | - Giancarlo Pesce
- Department of Diagnostic and Public Health, Unit of Epidemiology and Medical Statistics, University of Verona, Piazzale L.A. Scuro, 10 - 37134, Verona, Italy
| | - Anna Maria Fratta Pasini
- Department of Medicine, Section of Internal Medicine, University of Verona, Piazzale L.A. Scuro, 10 - 37134, Verona, Italy
| | - Luciano Cominacini
- Department of Medicine, Section of Internal Medicine, University of Verona, Piazzale L.A. Scuro, 10 - 37134, Verona, Italy
| |
Collapse
|
20
|
Santoro F, Costantino MD, Guastafierro F, Triggiani G, Ferraretti A, Tarantino N, Saguner A, Di Biase M, Brunetti ND. Inflammatory patterns in Takotsubo cardiomyopathy and acute coronary syndrome: A propensity score matched analysis. Atherosclerosis 2018; 274:157-161. [DOI: 10.1016/j.atherosclerosis.2018.05.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 05/06/2018] [Accepted: 05/10/2018] [Indexed: 01/05/2023]
|
21
|
Ueland T, Kleveland O, Michelsen AE, Wiseth R, Damås JK, Holven KB, Aukrust P, Gullestad L, Yndestad A, Halvorsen B. Serum lipoprotein(a) is not modified by interleukin-6 receptor antagonism or associated with inflammation in non-ST-elevation myocardial infarction. Int J Cardiol 2018; 274:348-350. [PMID: 29961573 DOI: 10.1016/j.ijcard.2018.06.093] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/02/2018] [Accepted: 06/21/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND The IL-6 receptor antagonist tocilizumab has been shown to attenuate the proatherogenic lipoprotein a [Lp(a)] in rheumatoid arthritis. We evaluated if a single dose of tocilizumab reduced Lp(a) in patients with non-ST-elevation myocardial infarction (NSTEMI). METHODS Lp(a) was assessed by immunoassay (n = 117 patients) at 7 consecutive time-points between day 1 and 3 and at 3 and 6 months follow-up. RESULTS Tocilizumab did not affect Lp(a) at any time-point during the study and was not associated with cardiovascular risk factors. CONCLUSIONS Short-time inhibition of IL-6 with tocilizumab in patients with NSTEMI did not influence Lp(a) levels.
Collapse
Affiliation(s)
- Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; Institute of Clinical Medicine, Oslo University Hospital Rikshospitalet, Norway; K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway.
| | - Ola Kleveland
- Clinic of Cardiology, St. Olavs Hospital, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology NTNU, Trondheim, Norway
| | - Annika E Michelsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; Institute of Clinical Medicine, Oslo University Hospital Rikshospitalet, Norway
| | - Rune Wiseth
- Clinic of Cardiology, St. Olavs Hospital, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology NTNU, Trondheim, Norway
| | - Jan Kristian Damås
- Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology NTNU, Trondheim, Norway
| | - Kirsten B Holven
- Norwegian National Advisory Unit on Familial Hypercholesterolemia, Department of Endocrinology, Morbid Obesity, and Preventive Medicine, Norway; Department of Nutrition, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; Institute of Clinical Medicine, Oslo University Hospital Rikshospitalet, Norway; Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Norway; K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway
| | - Lars Gullestad
- Institute of Clinical Medicine, Oslo University Hospital Rikshospitalet, Norway; Department of Cardiology, Oslo University Hospital Rikshospitalet, Norway; KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Norway
| | - Arne Yndestad
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; Institute of Clinical Medicine, Oslo University Hospital Rikshospitalet, Norway; KG Jebsen Center for Cardiac Research, University of Oslo and Center for Heart Failure Research, Oslo University Hospital, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; Institute of Clinical Medicine, Oslo University Hospital Rikshospitalet, Norway
| |
Collapse
|
22
|
Abstract
Tocilizumab (TCZ) is an important biologic response modifier that rheumatologists routinely employ in the treatment of several systemic autoimmune diseases. TCZ binds to interleukin (IL)-6 receptors, inhibits cellular activation, and mitigates inflammation by IL-6. In mid-2017, TCZ was approved by the US Food and Drug Administration for its first nonrheumatologic condition, the treatment of chimeric antigen receptor (CAR) T cell-induced severe or life-threatening cytokine release syndrome in patients 2 years of age or older. With this approval and with the increasing use of TCZ off-label for other non-rheumatologic conditions such as Castleman's Disease and its variant TAFRO syndrome, where else might TCZ be successfully utilized as treatment? Recently interesting data has been published regarding possible use of TCZ in the treatment of myocardial infarction. This review focuses on the role of IL-6 and its receptor in myocardial inflammation and association with adverse clinical outcomes. Discussed are one animal study and two human trials that have been published studying the effect of TCZ in patients with acute myocardial infarction. Finally, this review summarizes the current data and makes recommendations for future clinical trial development in what hopefully will be a promising application of TCZ for a serious nonrheumatologic condition.
Collapse
Affiliation(s)
- Matthew B Carroll
- a Rheumatology, Keesler Medical Center, Keesler AFB , Biloxi , MS , USA
| |
Collapse
|
23
|
Sharif K, Watad A, Bragazzi NL, Asher E, Abu Much A, Horowitz Y, Lidar M, Shoenfeld Y, Amital H. Anterior ST-elevation myocardial infarction induced by rituximab infusion: A case report and review of the literature. J Clin Pharm Ther 2018; 42:356-362. [PMID: 28440561 DOI: 10.1111/jcpt.12522] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 03/05/2017] [Indexed: 12/23/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVES Rituximab is a chimeric monoclonal anti-CD20 antibody approved for the treatment of some lymphoid malignancies as well as for autoimmune diseases including rheumatoid arthritis (RA), idiopathic thrombocytopenic purpura (ITP) and vasculitis. Generally, rituximab is well tolerated; nevertheless, some patients develop adverse effects including infusion reactions. Albeit rare, these reactions may in some cases be life-threatening conditions. Rituximab cardiovascular side effects include more common effects such as hypertension, oedema and rare cases of arrhythmias and myocardial infarction. CASE SUMMARY In this article, we report a case of a 58-year-old man with a history of overlap syndrome including RA and limited scleroderma who was treated with rituximab and developed a dramatic ST-elevation myocardial infarction (STEMI) during the drug administration. WHAT IS NEW AND CONCLUSION This report underlines previous published reports emphasizing the awareness of such an association. This communication also warrants the importance of screening for ischaemic heart disease in selected cases of patients treated with rituximab.
Collapse
Affiliation(s)
- K Sharif
- Department of Medicine "B", Sheba Medical Center, Tel-Hashomer, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Hashomer, Israel
| | - A Watad
- Department of Medicine "B", Sheba Medical Center, Tel-Hashomer, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Hashomer, Israel.,Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
| | - N L Bragazzi
- School of Public Health, Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - E Asher
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Hashomer, Israel.,The Leviev Heart Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - A Abu Much
- Department of Medicine "B", Sheba Medical Center, Tel-Hashomer, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Hashomer, Israel.,The Leviev Heart Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - Y Horowitz
- Department of Medicine "B", Sheba Medical Center, Tel-Hashomer, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Hashomer, Israel
| | - M Lidar
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Hashomer, Israel.,Rheumatology Unit, Sheba Medical Center, Tel-Hashomer, Israel
| | - Y Shoenfeld
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Hashomer, Israel.,Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
| | - H Amital
- Department of Medicine "B", Sheba Medical Center, Tel-Hashomer, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Hashomer, Israel.,Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
| |
Collapse
|
24
|
Larsen SB, Grove EL, Würtz M, Neergaard-Petersen S, Hvas AM, Kristensen SD. The influence of low-grade inflammation on platelets in patients with stable coronary artery disease. Thromb Haemost 2017; 114:519-29. [DOI: 10.1160/th14-12-1007] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 05/07/2015] [Indexed: 12/19/2022]
Abstract
SummaryInflammation is likely to be involved in all stages of atherosclerosis. Numerous inflammatory biomarkers are currently being studied, and even subtle increases in inflammatory biomarkers have been associated with increased risk of cardiovascular events in patients with coronary artery disease (CAD). Low-grade inflammation may influence both platelet production and platelet activation potentially leading to enhanced platelet aggregation. Thrombopoietin is considered the primary regulator of platelet production, but it likely acts in conjunction with numerous cytokines, of which many have altered levels in CAD. Previous studies have shown that high-sensitive C-reactive protein (CRP) independently predicts increased platelet aggregation in stable CAD patients. Increased levels of CRP, fibrinogen, interleukin-6, stromal cell-derived factor-1, CXC motif ligand 16, macrophage migration inhibitory factor, RANTES, calprotectin, and copeptin have been associated with increased risk of cardiovascular events in CAD patients. Additionally, some of these inflammatory markers have been associated with enhanced platelet activation and aggregation. However, CRP and other inflammatory markers provide only limited additional predictive value to classical risk factors such as smoking, blood pressure, and cholesterol levels. Existing data do not clarify whether inflammation simply accompanies CAD and increased production and aggregation of platelets, or whether a causal relationship exists. In this review, we provide a comprehensive overview of inflammatory markers in stable CAD with particular emphasis on platelet production, activation, and aggregation in CAD patients.
Collapse
|
25
|
Cho JY. Identification of Risk Factors Influencing In-Stent Restenosis with Acute Coronary Syndrome Presentation. Chonnam Med J 2017; 53:203-210. [PMID: 29026708 PMCID: PMC5636759 DOI: 10.4068/cmj.2017.53.3.203] [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: 06/22/2017] [Revised: 07/22/2017] [Accepted: 08/02/2017] [Indexed: 01/22/2023] Open
Abstract
Although the angiographic rates of in-stent restenosis (ISR) at later months have reduced dramatically with the introduction of drug-eluting stents (DESs), some patients with ISR after implantation of DES present with acute coronary syndrome (ACS). Here, we sought to identify parameters influencing the likelihood of restenosis with ACS presentation after DES implantation. Stented patients (n=3,817) with DESs in the Korea University Anam Hospital percutaneous coronary intervention registry were reviewed retrospectively for inclusion. In this database, 247 age- and sex-matched patients (6.5%) with ISR were allocated to either the Stable ISR group (n=78) or the ACS ISR group (n=73). Predictors of in-stent restenosis were identified with Cox regression analyses. Age (hazard ratio [HR], 1.14; 95% confidence interval [CI], 1.02 to 1.27; p=0.026), diabetes (HR, 8.40; 95% CI, 1.30 to 54.1; p=0.025), use of aspirin (HR, 0.003; 95% CI, 0.0001 to 0.63; p=0.03), clopidogrel (HR, 0.005; 95% CI, 0.001 to 0.121; p=0.001), renin-angiotensin system (RAS) blocker (HR, 0.02; 95% CI, 0.003 to 0.14; p<0.001), use of first -generation DES (HR, 0.07; 95% CI, 0.009 to 0.59; p=0.014), and matrix metalloproteinase 2 (MMP-2) levels (HR, 1.120; 95% CI, 1.001 to 1.190; p=0.004) during follow-up angiograms were significant predictors of ISR with ACS presentation during the 3 year follow-up. Age, diabetes, the use of first generation DES, and increased MMP-2 levels were significant predictors of ISR with ACS presentation; moreover, the use of aspirin, clopidogrel, RAS blocker, and the use of second generation DESs prevented ISR with ACS presentation.
Collapse
Affiliation(s)
- Jae Young Cho
- Department of Cardiovascular Medicine, Regional Cardiocerebrovascular Center, Wonkang University Hospital, Iksan, Korea
| |
Collapse
|
26
|
Gargiulo S, Testa G, Gamba P, Staurenghi E, Poli G, Leonarduzzi G. Oxysterols and 4-hydroxy-2-nonenal contribute to atherosclerotic plaque destabilization. Free Radic Biol Med 2017; 111:140-150. [PMID: 28057601 DOI: 10.1016/j.freeradbiomed.2016.12.037] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 12/22/2016] [Accepted: 12/24/2016] [Indexed: 12/31/2022]
Abstract
A growing bulk of evidence suggests that cholesterol oxidation products, known as oxysterols, and 4-hydroxy-2-nonenal (HNE), the major proatherogenic components of oxidized low density lipoproteins (oxLDLs), significantly contribute to atherosclerotic plaque progression and destabilization, with eventual plaque rupture. These oxidized lipids are involved in various key steps of this complex process, mainly thanks to their ability to induce inflammation, oxidative stress, and apoptosis. This review summarizes the current knowledge of the effects induced by these compounds on vascular cells, after their accumulation in the arterial wall and in the atherosclerotic plaque.
Collapse
Affiliation(s)
- Simona Gargiulo
- Department of Clinical and Biological Sciences, University of Torino, San Luigi Hospital, 10043 Orbassano, Torino, Italy
| | - Gabriella Testa
- Department of Clinical and Biological Sciences, University of Torino, San Luigi Hospital, 10043 Orbassano, Torino, Italy
| | - Paola Gamba
- Department of Clinical and Biological Sciences, University of Torino, San Luigi Hospital, 10043 Orbassano, Torino, Italy
| | - Erica Staurenghi
- Department of Clinical and Biological Sciences, University of Torino, San Luigi Hospital, 10043 Orbassano, Torino, Italy
| | - Giuseppe Poli
- Department of Clinical and Biological Sciences, University of Torino, San Luigi Hospital, 10043 Orbassano, Torino, Italy
| | - Gabriella Leonarduzzi
- Department of Clinical and Biological Sciences, University of Torino, San Luigi Hospital, 10043 Orbassano, Torino, Italy.
| |
Collapse
|
27
|
Kitazawa F, Fuchida SI, Ise F, Kado Y, Ueda K, Kokufu T, Okano A, Hatsuse M, Murakami S, Nakayama Y, Takara K, Shimazaki C. Combination of lenalidomide and low-dose dexamethasone therapy promotes the anticoagulant activity of warfarin in patients with immunoglobulin light-chain amyloidosis. Oncol Lett 2017; 14:475-479. [DOI: 10.3892/ol.2017.6133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 02/23/2017] [Indexed: 11/05/2022] Open
|
28
|
Milić V, Džudović B, Obradović S. INFLUENCE OF SYSTEMIC INFLAMMATORY RESPONSE ON IN HOSPITAL OUTCOME IN PATIENTS WITH TYPE 2 DIABETES MELLITUS AND ST-SEGMENT ELEVATION MYOCARDIAL INFARCTION UNDERGOING PRIMARY PERCUTANEOUS CORONARY INTERVENTION. ACTA MEDICA MEDIANAE 2017. [DOI: 10.5633/amm.2017.0201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
29
|
Serum Cytokine Profile in Relation to the Severity of Coronary Artery Disease. BIOMED RESEARCH INTERNATIONAL 2017; 2017:4013685. [PMID: 28349060 PMCID: PMC5352875 DOI: 10.1155/2017/4013685] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 02/09/2017] [Accepted: 02/12/2017] [Indexed: 11/25/2022]
Abstract
Objectives. To investigate the potential association of a set of serum cytokines with the severity of coronary artery disease (CAD). Methods. A total of 201 patients who underwent coronary angiography for chest discomfort were enrolled. The concentrations of serum IFN-γ, TNF-α, IL-2, IL-4, IL-6, IL-10, IL-9, and IL-17 were determined by xMAP multiplex technology. The CAD severity was assessed by Gensini score (GS). Results. The serum levels of TNF-α, IL-6, IL-9, IL-10, and IL-17 were significantly higher in high GS group (GS ≥ 38.5) than those in low GS group (GS < 38.5). Positive correlations were also found between these cytokines and the severity of CAD. After adjustment for other associated factors, three serum cytokines (IL-6, IL-9, and IL-17) and two clinical risk factors (creatinine and LDL-C) were identified as the independent predictors of increased severity of CAD. ROC curve analysis revealed that the logistic regression risk prediction model had a good performance on predicting CAD severity. Conclusions. Combinatorial analysis of serum cytokines (IL-6, IL-9, and IL-17) with clinical risk factors (creatinine and LDL-C) may contribute to the evaluation of the severity of CAD and may help guide the risk stratification of angina patients, especially in primary health facilities and in the catheter lab resource-limited settings.
Collapse
|
30
|
Neutrophil-derived microparticles are released into the coronary circulation following percutaneous coronary intervention in acute coronary syndrome patients. Biosci Rep 2017; 37:BSR20160430. [PMID: 27913753 PMCID: PMC5240586 DOI: 10.1042/bsr20160430] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 11/20/2016] [Accepted: 12/01/2016] [Indexed: 11/17/2022] Open
Abstract
To evaluate (i) local coronary and systemic levels of microparticles (MP) in acute coronary syndrome (ACS) and stable angina pectoris (SAP) patients and (ii) their release after plaque disruption with percutaneous coronary intervention (PCI). MP are small vesicles originating from plasma membranes of cells after activation or apoptosis and are implicated in the pathogenesis of atherosclerosis. Neutrophils play a role in plaque destabilization and shed neutrophil-derived MP that have the potential to drive significant proinflammatory and thrombotic downstream effects. Eight ACS and eight SAP patients were included. Coronary sinus (CS) samples pre-intervention (CS1), 45 s following balloon angioplasty (CS2) and at 45 s intervals following stent deployment (CS3, CS4 and CS5), together with peripheral vein samples, pre- and post-PCI were analysed for neutrophil-derived (CD66b+), endothelial-derived (CD144+), platelet-derived (CD41a+), monocyte-derived (CD14+) and apoptotic (Annexin V+) MP. ELISA for interleukin (IL)-6, myeloperoxidase (MPO) and P-selectin was also performed. CD66b+ MP levels were similar in both groups pre-intervention. Post-PCI, CS levels rose significantly in ACS but not SAP patients (ACS area under the curve (AUC): 549 ± 83, SAP AUC: 24 ± 29, P<0.01). CS CD41a+, CD144+, CD14+ and Annexin V+ MP levels did not differ between groups. Acute neutrophil-derived MP release post-PCI occurs in ACS compared with stable patients, likely to be reflective of plaque MP content in vulnerable lesions.
Collapse
|
31
|
Gliemann L, Nyberg M, Hellsten Y. Effects of exercise training and resveratrol on vascular health in aging. Free Radic Biol Med 2016; 98:165-176. [PMID: 27085843 DOI: 10.1016/j.freeradbiomed.2016.03.037] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 03/16/2016] [Accepted: 03/30/2016] [Indexed: 01/06/2023]
Abstract
Cardiovascular disease is a leading cause of death in the western world with aging being one of the strongest predictors of cardiovascular events. Aging is associated with impaired vascular function due to endothelial dysfunction and altered redox balance, partly caused by an increased formation of reactive oxygen species combined with a reduction in the endogenous antioxidant capacity. The consequence of these alterations is a reduced bioavailability of nitric oxide (NO) with implications for aspects such as control of vascular tone and low grade inflammation. However, it is not only aging per se but also the accumulative influence of physical inactivity and other life-style factors, which negatively affect the vascular system. Regular physical activity improves NO bioavailability, the redox balance and the plasma lipid profile and, at a functional level, reduces or even reverses a majority of the observed detrimental effects of aging on vascular function. The effects of aging and physical activity on vascular function are, in part, related to alterations in cellular signaling through sirtuin-1, AMPK and the estrogen receptor. The polyphenol resveratrol can activate these same pathways and has, in animals and in vitro models, been shown to act as a partial mimetic of physical activity. However, support for beneficial effects of resveratrol in human is weak and studies even show that resveratrol supplementation, similarly to supplementation with other antioxidants, can counteract the positive effects of physical activity. Regular physical activity remains the most effective way of maintaining and improving vascular health status and caution should be taken regarding potential interference of supplements on training adaptations.
Collapse
Affiliation(s)
- Lasse Gliemann
- Department of Nutrition, Exercise and Sports, Section for Integrative Physiology, University of Copenhagen, Universitetsparken 13, DK-2100 Copenhagen Ø, Denmark
| | - Michael Nyberg
- Department of Nutrition, Exercise and Sports, Section for Integrative Physiology, University of Copenhagen, Universitetsparken 13, DK-2100 Copenhagen Ø, Denmark
| | - Ylva Hellsten
- Department of Nutrition, Exercise and Sports, Section for Integrative Physiology, University of Copenhagen, Universitetsparken 13, DK-2100 Copenhagen Ø, Denmark.
| |
Collapse
|
32
|
Čermák T, MuŽáková V, Matějka J, Skalický J, Laštovička P, Líbalová M, Kanďár R, Novotný V, Čegan A. Fatty acid profile in erythrocyte membranes and plasma phospholipids affects significantly the extent of inflammatory response to coronary stent implantation. Physiol Res 2016; 65:941-951. [PMID: 27539101 DOI: 10.33549/physiolres.933194] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
In coronary heart disease, the treatment of significant stenosis by percutaneous coronary intervention (PCI) with stent implantation elicits local and systemic inflammatory responses. This study was aimed at evaluation of the dynamics of inflammatory response and elucidation of the relationship between the fatty acid profile of red blood cell (RBC) membranes or plasma phospholipids and inflammation after PCI. High-sensitivity C-reactive protein (hsCRP), interleukin-6 (IL-6), serum amyloid A (SAA), malondialdehyde (MDA) and the fatty acid profiles were determined in patients with advanced coronary artery disease undergoing PCI before, 24 h and 48 h after drug-eluting stent implantation (n=36). Patients after PCI exhibited a significant increase in studied markers (hsCRP, IL-6, SAA, MDA). Many significant associations were found between the increase of IL-6, resp. SAA and the amounts of n-6 polyunsaturated fatty acids (namely linoleic, dihomo-gamma-linolenic, docosatetraenoic and docosapentaenoic acid), resp. saturated fatty acids (pentadecanoic, stearic, nonadecanoic) in erythrocyte membranes. The magnitude of the inflammatory response to PCI is related to erythrocyte membrane fatty acid profile, which seems to be a better potential predictor of elevation of inflammatory markers after PCI than plasma phospholipids.
Collapse
Affiliation(s)
- T Čermák
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Pardubice, Czech Republic.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Riccioni G, Gammone MA, Tettamanti G, Bergante S, Pluchinotta FR, D'Orazio N. Resveratrol and anti-atherogenic effects. Int J Food Sci Nutr 2015; 66:603-10. [PMID: 26306466 DOI: 10.3109/09637486.2015.1077796] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The role of inflammation and oxidative stress in atherosclerosis development has been increasingly well recognized over the past decade. Inflammation has a significant role at all stages of atherosclerosis, including initiation, progression and plaque formation. Resveratrol is a naturally occurring polyphenolic compound found in grape products, berry fruits and red wine. Its ability to behave therapeutically as a component of red wine has attracted wide attention. Accumulating evidence suggests that it is a highly pleiotropic molecule that modulates numerous targets and molecular functions. Epidemiological studies indicate that the Mediterranean diet, rich in resveratrol, is associated with a reduced risk of atherosclerosis. Resveratrol is believed to decrease circulating low-density lipoprotein cholesterol levels, reduce cardiovascular disease risk; it reduces lipid peroxidation, platelet aggregation and oxidative stress. Resveratrol is considered a safe compound, since no significant toxic effects have been demonstrated after administration of a broad range of concentrations, and an effective anti-atherogenic agent.
Collapse
Affiliation(s)
- Graziano Riccioni
- a Cardiology Unit, San Camillo de Lellis Hospital , Manfredonia , Foggia , Italy
| | - Maria Alessandra Gammone
- b Clinical and Human Nutrition Unit, Department of Oral Medical and Biotechnological Sciences, University "G. D'Annunzio" , Chieti , Italy and
| | - Guido Tettamanti
- c Department of Pediatric Cardiovascular Surgery , IRCCS San Donato Milanese Hospital , San Donato Milanese , Italy
| | - Sonia Bergante
- c Department of Pediatric Cardiovascular Surgery , IRCCS San Donato Milanese Hospital , San Donato Milanese , Italy
| | - Francesca Romana Pluchinotta
- c Department of Pediatric Cardiovascular Surgery , IRCCS San Donato Milanese Hospital , San Donato Milanese , Italy
| | - Nicolantonio D'Orazio
- b Clinical and Human Nutrition Unit, Department of Oral Medical and Biotechnological Sciences, University "G. D'Annunzio" , Chieti , Italy and
| |
Collapse
|
34
|
Martínez GJ, Robertson S, Barraclough J, Xia Q, Mallat Z, Bursill C, Celermajer DS, Patel S. Colchicine Acutely Suppresses Local Cardiac Production of Inflammatory Cytokines in Patients With an Acute Coronary Syndrome. J Am Heart Assoc 2015; 4:e002128. [PMID: 26304941 PMCID: PMC4599469 DOI: 10.1161/jaha.115.002128] [Citation(s) in RCA: 186] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 06/26/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Interleukin (IL)-1β, IL-18, and downstream IL-6 are key inflammatory cytokines in the pathogenesis of coronary artery disease. Colchicine is believed to block the NLRP3 inflammasome, a cytosolic complex responsible for the production of IL-1β and IL-18. In vivo effects of colchicine on cardiac cytokine release have not been previously studied. This study aimed to (1) assess the local cardiac production of inflammatory cytokines in patients with acute coronary syndromes (ACS), stable coronary artery disease and in controls; and (2) determine whether acute administration of colchicine inhibits their production. METHODS AND RESULTS Forty ACS patients, 33 with stable coronary artery disease, and 10 controls, were included. ACS and stable coronary artery disease patients were randomized to oral colchicine treatment (1 mg followed by 0.5 mg 1 hour later) or no colchicine, 6 to 24 hours prior to cardiac catheterization. Blood samples from the coronary sinus, aortic root (arterial), and lower right atrium (venous) were collected and tested for IL-1β, IL-18, and IL-6 using ELISA. In ACS patients, coronary sinus levels of IL-1β, IL-18, and IL-6 were significantly higher than arterial and venous levels (P=0.017, <0.001 and <0.001, respectively). Transcoronary (coronary sinus-arterial) gradients for IL-1β, IL-18, and IL-6 were highest in ACS patients and lowest in controls (P=0.077, 0.033, and 0.014, respectively). Colchicine administration significantly reduced transcoronary gradients of all 3 cytokines in ACS patients by 40% to 88% (P=0.028, 0.032, and 0.032, for IL-1β, IL-18, and IL-6, respectively). CONCLUSIONS ACS patients exhibit increased local cardiac production of inflammatory cytokines. Short-term colchicine administration rapidly and significantly reduces levels of these cytokines.
Collapse
Affiliation(s)
- Gonzalo J Martínez
- Department of Cardiology, Royal Prince Alfred HospitalSydney, New South Wales, Australia
- Sydney Medical School, The University of SydneyNew South Wales, Australia
- Department of Cardiology, Catholic University School of MedicineSantiago, Chile
| | - Stacy Robertson
- Sydney Medical School, The University of SydneyNew South Wales, Australia
- Heart Research InstituteSydney, New South Wales, Australia
| | - Jennifer Barraclough
- Department of Cardiology, Royal Prince Alfred HospitalSydney, New South Wales, Australia
| | - Qiong Xia
- Sydney Medical School, The University of SydneyNew South Wales, Australia
| | - Ziad Mallat
- Institut National de la Santé et de la Recherche Médicale (INSERM), Paris-Cardiovascular Research CenterParis, France
- Division of Cardiovascular Medicine, Addenbrooke’s Hospital, University of CambridgeUnited Kingdom
| | - Christina Bursill
- Sydney Medical School, The University of SydneyNew South Wales, Australia
- Heart Research InstituteSydney, New South Wales, Australia
| | - David S Celermajer
- Department of Cardiology, Royal Prince Alfred HospitalSydney, New South Wales, Australia
- Sydney Medical School, The University of SydneyNew South Wales, Australia
- Heart Research InstituteSydney, New South Wales, Australia
| | - Sanjay Patel
- Department of Cardiology, Royal Prince Alfred HospitalSydney, New South Wales, Australia
- Sydney Medical School, The University of SydneyNew South Wales, Australia
- Heart Research InstituteSydney, New South Wales, Australia
| |
Collapse
|
35
|
Insufficient glucocorticoid signaling and elevated inflammation in coronary heart disease patients with comorbid depression. Brain Behav Immun 2015; 48:8-18. [PMID: 25683698 DOI: 10.1016/j.bbi.2015.02.002] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 01/19/2015] [Accepted: 02/03/2015] [Indexed: 12/23/2022] Open
Abstract
Coronary heart disease (CHD) and depression are very common and often co-existing disorders. In addition to psychological and social morbidity, depression exacerbates adverse cardiac outcomes in CHD patients. Inflammation has been proposed as one of the mechanisms involved in the association between these two debilitating diseases. Therefore, the present study aimed to evaluate inflammatory responses as well as to investigate the pathophysiological mechanisms underlying the putative inflammatory activation in CHD patients with and without depression, by assessing the function of two important biological factors regulating inflammation, the hypothalamus-pituitary-adrenal (HPA) axis and the glucocorticoid receptor (GR). Eighty-three CHD patients with (n=28) and without (n=55) comorbid depression were recruited from primary care services in South London. Depression status was assessed by means of Clinical Interview Schedule Revised for diagnosis of depression, and Beck Depression Inventory for the presence of depressive symptoms. Serum C-reactive protein (CRP), plasma vascular endothelial growth factor (VEGF), and plasma and salivary cortisol were measured using commercially available ELISA kits. Gene expression of GR and interleukin-6 (IL-6) were conducted via qPCR. GR sensitivity was evaluated in vitro in isolated peripheral blood mononuclear cells using the dexamethasone inhibition of lipopolysaccharide-stimulated IL-6 levels. Serum levels of kynurenine pathway metabolites were measured using high performance liquid chromatography. Our results show that CHD patients with depression had higher levels of CRP, IL-6 gene expression, and VEGF compared with CHD non-depressed, as well as lower plasma and saliva cortisol levels. The CHD depressed group also exhibited a reduction in GR expression and sensitivity. Finally, tryptophan levels were significantly lower in patients with depression, who also showed an increased kynurenine/tryptophan ratio. In conclusion, CHD patients with depression had elevated levels of inflammation in the context of HPA axis hypoactivity, GR resistance, and increased activation of the kynurenine pathway. Reduced cortisol bioavailability and attenuated glucocorticoid responsiveness due to decreased expression and sensitivity of GR may lead to insufficient glucocorticoid signaling and thus elevation of inflammation in these patients.
Collapse
|
36
|
Koyama K, Yoneyama K, Mitarai T, Ishibashi Y, Takahashi E, Kongoji K, Harada T, Akashi YJ. Association between inflammatory biomarkers and thin-cap fibroatheroma detected by optical coherence tomography in patients with coronary heart disease. Arch Med Sci 2015; 11:505-12. [PMID: 26170842 PMCID: PMC4495146 DOI: 10.5114/aoms.2015.52352] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Revised: 06/07/2014] [Accepted: 06/15/2014] [Indexed: 01/03/2023] Open
Abstract
INTRODUCTION The relationship between plaque morphology detected by optical coherence tomography (OCT) and inflammatory biomarkers is not well known. MATERIAL AND METHODS This study included 47 patients with ischemic heart disease (22 patients with acute coronary syndrome and 25 patients with effort angina pectoris) who underwent percutaneous coronary intervention (PCI). Before PCI, peripheral blood levels of the inflammatory biomarkers high-sensitivity C-reactive protein (hs-CRP) and interleukin-6 (IL-6) were measured. The OCT can detect thin-cap fibroatheroma (TCFA), a lesion with high potential for adverse cardiac events. We investigated the relationships between TCFAs in culprit lesions detected by OCT and the peripheral blood levels of these biomarkers. RESULTS We observed 12 lesions detected as TCFAs. The natural logs of hs-CRP and IL-6 levels in the TCFA group were higher than those in the non-TCFA group (hs-CRP 0.87 (-0.96 to 0.87) vs. -0.47 (-0.92 to 0.30) mg/l, p = 0.027; and IL-6 1.63 (0.63-3.23) vs. 0.53 (-0.21 to 1.05) pg/dl, p = 0.005, respectively). In multivariate logistic regression analysis, log IL-6 was an independent predictor for TCFA detected by OCT (log IL-6, 0.970 pg/dl, p = 0.023). Receiver operating characteristic curve analysis confirmed that IL-6, compared to hs-CRP, has a higher area under the curve for predicting TCFA (0.783 vs. 0.715, respectively). CONCLUSIONS Peripheral blood levels of both hs-CRP and IL-6 were associated with TCFAs, as detected by OCT. Moreover, IL-6 has a higher potential than hs-CRP for predicting TCFA.
Collapse
Affiliation(s)
- Kohei Koyama
- Division of Cardiology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Kihei Yoneyama
- Division of Cardiology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Takanobu Mitarai
- Division of Cardiology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Yuki Ishibashi
- Division of Cardiology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Eiji Takahashi
- St. Marianna University School of Medicine, Yokohama-city Seibu Hospital, Yokohama, Japan
| | - Ken Kongoji
- Division of Cardiology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Tomoo Harada
- Division of Cardiology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Yoshihiro J. Akashi
- Division of Cardiology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| |
Collapse
|
37
|
Spoto B, Mattace-Raso F, Sijbrands E, Leonardis D, Testa A, Pisano A, Pizzini P, Cutrupi S, Parlongo RM, D'Arrigo G, Tripepi G, Mallamaci F, Zoccali C. Association of IL-6 and a functional polymorphism in the IL-6 gene with cardiovascular events in patients with CKD. Clin J Am Soc Nephrol 2014; 10:232-40. [PMID: 25492254 DOI: 10.2215/cjn.07000714] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND OBJECTIVES High serum IL-6 is a major risk factor for cardiovascular disease (CVD) in the general population. This cytokine is substantially increased in patients with CKD, but it is still unknown whether the link between IL-6 and CVD in CKD is causal in nature. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS In a cohort of 755 patients with stages 2-5 CKD, consecutively recruited from 22 nephrology units in southern Italy, this study assessed the relationship of serum IL-6 with history of CVD, as well as with incident cardiovascular (CV) events (mean follow up±SD, 31±10 months) and used the functional polymorphism (-174 G/C) in the promoter of the IL-6 gene to investigate whether the link between IL-6 and CV events is causal. RESULTS In adjusted analyses, serum IL-6 above the median value was associated with history of CVD (P<0.001) and predicted the incidence rate of CV events (hazard ratio, 1.66; 95% confidence interval [95% CI], 1.11 to 2.49; P=0.01). Patients homozygous for the risk allele (C) of the -174 G/C polymorphism had higher levels of IL-6 than did those with other genotypes (P=0.04). Homozygous CC patients more frequently had a history of CVD (odds ratio, 2.15; 95% CI, 1.15 to 4.00; P=0.02) as well as a 87% higher rate of incident CV events (hazard ratio, 1.87; 95% CI, 1.02 to 3.44; P=0.04) compared with other genotypes. CONCLUSIONS In patients with stages 2-5 CKD, high serum IL-6 is associated with history of CVD and predicts incident CV events. The parallel relationship with history of CVD and incident CV events of the -174 G/C polymorphism in the IL-6 gene suggests that IL-6 may be causally involved in the high CV risk in this population.
Collapse
Affiliation(s)
- Belinda Spoto
- National Research Council of Italy-Institute of Clinical Physiology & Institute of Biomedicine and Molecular Immunology & Nephrology, Dialysis and Transplantation Unit of Reggio, Calabria, Italy; and
| | - Francesco Mattace-Raso
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Eric Sijbrands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Daniela Leonardis
- National Research Council of Italy-Institute of Clinical Physiology & Institute of Biomedicine and Molecular Immunology & Nephrology, Dialysis and Transplantation Unit of Reggio, Calabria, Italy; and
| | - Alessandra Testa
- National Research Council of Italy-Institute of Clinical Physiology & Institute of Biomedicine and Molecular Immunology & Nephrology, Dialysis and Transplantation Unit of Reggio, Calabria, Italy; and
| | - Anna Pisano
- National Research Council of Italy-Institute of Clinical Physiology & Institute of Biomedicine and Molecular Immunology & Nephrology, Dialysis and Transplantation Unit of Reggio, Calabria, Italy; and
| | - Patrizia Pizzini
- National Research Council of Italy-Institute of Clinical Physiology & Institute of Biomedicine and Molecular Immunology & Nephrology, Dialysis and Transplantation Unit of Reggio, Calabria, Italy; and
| | - Sebastiano Cutrupi
- National Research Council of Italy-Institute of Clinical Physiology & Institute of Biomedicine and Molecular Immunology & Nephrology, Dialysis and Transplantation Unit of Reggio, Calabria, Italy; and
| | - Rosa M Parlongo
- National Research Council of Italy-Institute of Clinical Physiology & Institute of Biomedicine and Molecular Immunology & Nephrology, Dialysis and Transplantation Unit of Reggio, Calabria, Italy; and
| | - Graziella D'Arrigo
- National Research Council of Italy-Institute of Clinical Physiology & Institute of Biomedicine and Molecular Immunology & Nephrology, Dialysis and Transplantation Unit of Reggio, Calabria, Italy; and
| | - Giovanni Tripepi
- National Research Council of Italy-Institute of Clinical Physiology & Institute of Biomedicine and Molecular Immunology & Nephrology, Dialysis and Transplantation Unit of Reggio, Calabria, Italy; and
| | - Francesca Mallamaci
- National Research Council of Italy-Institute of Clinical Physiology & Institute of Biomedicine and Molecular Immunology & Nephrology, Dialysis and Transplantation Unit of Reggio, Calabria, Italy; and
| | - Carmine Zoccali
- National Research Council of Italy-Institute of Clinical Physiology & Institute of Biomedicine and Molecular Immunology & Nephrology, Dialysis and Transplantation Unit of Reggio, Calabria, Italy; and
| |
Collapse
|
38
|
Pan S, Wu D, Teschendorff AE, Hong T, Wang L, Qian M, Wang C, Wang X. JAK2-centered interactome hotspot identified by an integrative network algorithm in acute Stanford type A aortic dissection. PLoS One 2014; 9:e89406. [PMID: 24586754 PMCID: PMC3933461 DOI: 10.1371/journal.pone.0089406] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Accepted: 01/20/2014] [Indexed: 12/18/2022] Open
Abstract
The precise mechanisms underlying dissections, especially those without connective tissue diseases or congenital vascular diseases, are incompletely understood. This study attempted to identify both the expression profile of the dissected ascending aorta and the interactome hotspots associated with the disease, using microarray technology and gene regulatory network analysis. There were 2,737 genes differentially expressed between patients with acute Stanford type A aortic dissection and controls. Eight interactome hotspots significantly associated with aortic dissection were identified by an integrative network algorithm. In particular, we identified a JAK2-centered expression module, which was validated in an independent gene expression microarray data set, and which was characterized by over-expressed cytokines and receptors in acute aortic dissection cases, indicating that JAK2 may play a key role in the inflammatory process, which potentially contributes to the occurrence of acute aortic dissection. Overall, the analytical strategy used in this study offered the possibility to identify functional relevant network modules and subsequently facilitated the biological interpretation in the complicated disease.
Collapse
Affiliation(s)
- Sun Pan
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Duojiao Wu
- Biomedical Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Andrew E. Teschendorff
- UCL Cancer Institute, University College London, London, United Kingdom
- CAS-MPG Partner Institute for Computational Biology, Shanghai, China
| | - Tao Hong
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Linyan Wang
- Biomedical Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mengjia Qian
- Biomedical Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chunsheng Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- * E-mail: (CW); (XW)
| | - Xiangdong Wang
- Biomedical Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
- * E-mail: (CW); (XW)
| |
Collapse
|
39
|
Nasiripour S, Gholami K, Mousavi S, Mohagheghi A, Radfar M, Abdollahi M, Khazaeipour Z, Mojtahedzadeh M. Comparison of the Effects of Enoxaparin and Heparin on Inflammatory Biomarkers in Patients with ST-segment Elevated Myocardial Infarction: A prospective Open Label Pilot Clinical Trial. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2014; 13:583-90. [PMID: 25237354 PMCID: PMC4157034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Heparin and enoxaparin possess anti-inflammatory properties. We compared the effects of these drugs on inflammatory biomarkers in patients with ST-segment Elevated Myocardial Infarction (STEMI). Thirty four patients with STEMI randomly separated in two groups and received standard doses of heparin and enoxaparin. The serum concentration of Serum Amyloid A (SAA), C-Reactive Protein (CRP), Interleukin (IL)-6, ferritin and Myeloperoxidase (MPO) were measured at baseline, 12 ,24 and 48 hours after drug administration. SERUM CONCENTRATIONS OF SAA (P 0.02), CRP (P: 0.02) and ferritin (P: 0.01) significantly reduced in heparin group during measurements compared to baseline, circulating levels of IL-6 (P: 0.002), SAA (P: 0.009), CRP (P: 0.01) were significantly decreased in enoxaparin group. The overall difference in inflammatory biomarkers between heparin and enoxaparin group was not significant. Both heparin and enoxaparine reduced serum levels of inflammatory biomarkers in patients with STEMI. This effect may provide additional clinical benefit of these drugs in the treatment of STEMI patients.
Collapse
Affiliation(s)
- Somayyeh Nasiripour
- Clinical Pharmacy Department, Faculty of Pharmacy, Tehran University of Medical Sciences and Health Services, Tehran, Iran.
| | - kheirollah Gholami
- Clinical Pharmacy Department, Faculty of Pharmacy, Tehran University of Medical Sciences and Health Services and Research Center for Rational Use of Drugs, Tehran, Iran.
| | - Sarah Mousavi
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Abbas Mohagheghi
- Department of Cardiology, Shariati Hospital, Tehran University of Medical Sciences, Tehran 14114, Iran.
| | - Mania Radfar
- Clinical Pharmacy Department, Faculty of Pharmacy, Tehran University of Medical Sciences and Health Services, Tehran, Iran.
| | - Mohammad Abdollahi
- Faculty of Pharmacy, and Pharmaceutical Sciences Research Centre, Tehran University of Medical Sciences, Tehran, Iran.
| | - Zahra Khazaeipour
- Brain and Spinal Cord Injury Research Center, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mojtaba Mojtahedzadeh
- Clinical Pharmacy Department, Faculty of Pharmacy, Tehran University of Medical Sciences and Health Services, Tehran, Iran.
- Faculty of Pharmacy, and Pharmaceutical Sciences Research Centre, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
40
|
Kim S, Sung J, Jung ES, Park HC, Lee H, Chin HJ, Kim DK, Kim YS, Han JS, Joo KW. Hemodynamic and biochemical benefits of the objective measurement of fluid status in hemodialysis patients. TOHOKU J EXP MED 2013; 228:125-33. [PMID: 22990525 DOI: 10.1620/tjem.228.125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Subtle fluid imbalance can cause poor clinical outcomes among hemodialysis patients. However, the traditional subjective assessment of fluid status may be inadequate. We evaluated whether the objective measurement and optimization of fluid status could be beneficial for hemodynamic and biochemical parameters in hemodialysis patients. We enrolled 120 hemodialysis patients, who were clinically euvolemic for at least 3 months. Based on the results of a body composition monitor, we divided the patients into the following two groups: the hyperhydrated group (post hemodialysis fluid overload ≥ 1.1 L) and the dehydrated group (post hemodialysis fluid overload < -1.1 L). We reduced the patient's body weight in the hyperhydrated group and raised the body weight in the dehydrated group towards normohydration (-1.1 L ≤ fluid overload < 1.1 L) for 16 weeks. Forty-four of 120 patients were in the hyperhydrated group, and 18 of 120 patients in the dehydrated group. After 16 weeks, systolic blood pressure and pulse pressure decreased in the hyperhydrated group, while there was no increase in blood pressure in the dehydrated group after the intervention. Serum levels of monocyte chemotactic protein-1, an inflammatory marker, gradually decreased in the hyperhydrated group, and serum adiponectin levels, an anti-atherogenic biomarker, increased in the two groups. We found that hyperhydrated patients contributed over 1/3 of the participants despite enrolling clinically euvolemic patients and that body composition monitor-guided optimization of body fluid status may lead to improvement of inflammatory markers and anti-atherogenic adipokines as well as hemodynamic parameters in hemodialysis patients.
Collapse
Affiliation(s)
- Sejoong Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Bazaz R, Marriott HM, Francis SE, Dockrell DH. Mechanistic links between acute respiratory tract infections and acute coronary syndromes. J Infect 2013; 66:1-17. [DOI: 10.1016/j.jinf.2012.09.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 09/22/2012] [Accepted: 09/26/2012] [Indexed: 12/27/2022]
|
42
|
IL-6 gene polymorphisms and CAD risk: a meta-analysis. Mol Biol Rep 2012; 40:2589-98. [PMID: 23242654 DOI: 10.1007/s11033-012-2345-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Accepted: 12/09/2012] [Indexed: 10/27/2022]
|
43
|
Funck-Jensen KL, Dalsgaard J, Grove EL, Hvas AM, Kristensen SD. Increased platelet aggregation and turnover in the acute phase of ST-elevation myocardial infarction. Platelets 2012; 24:528-37. [DOI: 10.3109/09537104.2012.738838] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
44
|
Interleukin-6 and E-selectin in acute coronary syndromes and stable angina pectoris. Herz 2012; 37:926-30. [DOI: 10.1007/s00059-012-3648-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 05/17/2012] [Accepted: 06/06/2012] [Indexed: 12/13/2022]
|
45
|
Effect of the intravascular low energy laser illumination during percutaneous coronary intervention on the inflammatory process in vascular wall. Lasers Med Sci 2012; 28:763-8. [PMID: 22733406 PMCID: PMC3637891 DOI: 10.1007/s10103-012-1142-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Accepted: 06/08/2012] [Indexed: 11/23/2022]
Abstract
The angioplasty procedure is associated with a release of numerous factors triggering the local inflammatory reaction in vascular wall and leading thus to the restenosis. In this study, we hypothesize that the low-energy laser irradiation may exert beneficial effect by limiting this process. A group of 101 subjects (75 men and 26 women, mean age: 59.1 ± 10.3) treated with percutaneous coronary intervention (PCI), were recruited to this study. While 52 patients (40 men and 12 women) were subjected to the intravascular low-energy laser irradiation (λ = 808 nm) of dilated lesion during the PCI, the remaining patients (35 men and 14 women) constituted the control group. The levels of interleukin 1β, 6 and 10 (IL 1β, IL 6 and IL 10) were measured immediately before the procedure, and then at the 6th, 12th hour as well as after 1 month following the PCI. Significantly lower levels of IL 1β and IL 6 in the irradiated group during each analysis after the procedure were observed. Moreover, significantly lower IL 10 level in irradiated group within 6 and 12 hours after PCI was observed. Irradiation of the lesion with low-energy laser radiation during the PCI procedure results in a decrease in the levels of pro-inflammatory IL 1β and IL 6 as well as in an increase in the levels of anti-inflammatory IL 10, which may result in decreased risk for restenosis.
Collapse
|
46
|
Potekhina AV, Arefieva TI, Krasnikova TL, Provatorov SI, Masenko VP, Osyaeva MK, Noeva EA. Changes in the concentration of monocytic chemotaxic protein-1 in patients with unstable angina treated with arixtra. Bull Exp Biol Med 2012; 150:656-8. [PMID: 22235409 DOI: 10.1007/s10517-011-1215-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The time course of inflammatory reaction markers in the blood of patients with unstable angina was studied during therapy including arixtra. Plasma concentration of monocytic chemotaxic protein-1 (MCP-1) decreased on days 2 and 3 in patients receiving arixtra and a trend to an increase in MCP-1 concentration was observed on day 7 after the drug was discontinued. After 1 month, MCP-1 level decreased in all patients. The concentration of highly sensitive C-reactive protein also decreased 1 month after the disease onset; no changes in the concentrations of IL-8 and IL-2 receptor α-subunit were detected during these periods. It seems that arixtra is characterized by an anti-inflammatory effect manifesting by reduction of plasma chemokine MCP-1 concentration.
Collapse
Affiliation(s)
- A V Potekhina
- Russian Research and Practical Centre of Cardiology, Moscow, Russia.
| | | | | | | | | | | | | |
Collapse
|
47
|
Leonarduzzi G, Gamba P, Gargiulo S, Biasi F, Poli G. Inflammation-related gene expression by lipid oxidation-derived products in the progression of atherosclerosis. Free Radic Biol Med 2012; 52:19-34. [PMID: 22037514 DOI: 10.1016/j.freeradbiomed.2011.09.031] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 09/16/2011] [Accepted: 09/24/2011] [Indexed: 12/31/2022]
Abstract
Vascular areas of atherosclerotic development persist in a state of inflammation, and any further inflammatory stimulus in the subintimal area elicits a proatherogenic response; this alters the behavior of the artery wall cells and recruits further inflammatory cells. In association with the inflammatory response, oxidative events are also involved in the development of atherosclerotic plaques. It is now unanimously recognized that lipid oxidation-derived products are key players in the initiation and progression of atherosclerotic lesions. Oxidized lipids, derived from oxidatively modified low-density lipoproteins (LDLs), which accumulate in the intima, strongly modulate inflammation-related gene expression, through involvement of various signaling pathways. In addition, considerable evidence supports a proatherogenic role of a large group of potent bioactive lipids called eicosanoids, which derive from oxidation of arachidonic acid, a component of membrane phospholipids. Of note, LDL lipid oxidation products might regulate eicosanoid production, modulating the enzymatic degradation of arachidonic acid by cyclooxygenases and lipoxygenases; these enzymes might also directly contribute to LDL oxidation. This review provides a comprehensive overview of current knowledge on signal transduction pathways and inflammatory gene expression, modulated by lipid oxidation-derived products, in the progression of atherosclerosis.
Collapse
|
48
|
Carter AM. Complement activation: an emerging player in the pathogenesis of cardiovascular disease. SCIENTIFICA 2012; 2012:402783. [PMID: 24278688 PMCID: PMC3820556 DOI: 10.6064/2012/402783] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 11/06/2012] [Indexed: 05/08/2023]
Abstract
A wealth of evidence indicates a fundamental role for inflammation in the pathogenesis of cardiovascular disease (CVD), contributing to the development and progression of atherosclerotic lesion formation, plaque rupture, and thrombosis. An increasing body of evidence supports a functional role for complement activation in the pathogenesis of CVD through pleiotropic effects on endothelial and haematopoietic cell function and haemostasis. Prospective and case control studies have reported strong relationships between several complement components and cardiovascular outcomes, and in vitro studies and animal models support a functional effect. Complement activation, in particular, generation of C5a and C5b-9, influences many processes involved in the development and progression of atherosclerosis, including promotion of endothelial cell activation, leukocyte infiltration into the extracellular matrix, stimulation of cytokine release from vascular smooth muscle cells, and promotion of plaque rupture. Complement activation also influences thrombosis, involving components of the mannose-binding lectin pathway, and C5b-9 in particular, through activation of platelets, promotion of fibrin formation, and impairment of fibrinolysis. The participation of the complement system in inflammation and thrombosis is consistent with the physiological role of the complement system as a rapid effector system conferring protection following vessel injury. However, in the context of CVD, these same processes contribute to development of atherosclerosis, plaque rupture, and thrombosis.
Collapse
Affiliation(s)
- Angela M. Carter
- Division of Epidemiology, Leeds Institute of Genetics, Health and Therapeutics, Faculty of Medicine and Health and the Multidisciplinary Cardiovascular Research Centre, University of Leeds, Clarendon Way, Leeds LS2 9JT, UK
- *Angela M. Carter:
| |
Collapse
|
49
|
Effects of selective H.E.L.P. LDL-apheresis on plasma inflammatory markers concentration in severe dyslipidemia: Implication for anti-inflammatory response. Cytokine 2011; 56:850-4. [DOI: 10.1016/j.cyto.2011.08.038] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Revised: 08/22/2011] [Accepted: 08/25/2011] [Indexed: 11/18/2022]
|
50
|
Abstract
The study was designed to assess the cardiac release kinetics of the cytokines interleukin-1ß (IL-1ß), interleukin-6 (IL-6) and tumor-necrosis-factor-α (TNF-α) in patients with significant stenosis of the ramus interventricularis anterior. Ten patients were treated by bare metal stent implantation, 11 patients who underwent a diagnostic coronary angiography without intervention served as a control group. Cytokines paired blood samples were withdrawn from the coronary sinus and a peripheral vein immediately before and 1, 2, 6 h after the intervention. Myocardial ischemia was monitored by means of cardiac lactate metabolism and 12-lead electrocardiogram. After coronary intervention IL-6 gradually increased from a common baseline level of 1.34 ± 1.56 pg/ml to a maximum of 10.58 ± 5.7 pg/ml in the peripheral vein and 15.81 ± 6.98 pg/ml in the coronary sinus within 6 h with persistent higher levels in the coronary sinus indicating coronary IL-6 release. After 12 h the peripheral venous concentration of IL-6 returned to baseline levels. Neither in the study group for IL-1ß and TNF-α nor in the control group for any cytokine level significant changes were found. Myocardial ischemia was excluded in all patients. Uncomplicated percutaneous coronary intervention (PCI) was followed by a significant cardiac IL-6 release due to endothelial injury and not to myocardial ischemia.
Collapse
|