1
|
Van Laecke S, Van Damme K, Dendooven A. Immunosenescence: an unexplored role in glomerulonephritis. Clin Transl Immunology 2022; 11:e1427. [PMID: 36420421 PMCID: PMC9676375 DOI: 10.1002/cti2.1427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/15/2022] [Accepted: 10/16/2022] [Indexed: 11/22/2022] Open
Abstract
Immunosenescence is a natural ageing phenomenon with alterations in innate and especially adaptive immunity and contributes to reduced antimicrobial defence and chronic low‐grade inflammation. This is mostly reflected by an increase in organ‐directed and/or circulating reactive and cytolytic terminally differentiated T cells that have lost their expression of the costimulatory receptor CD28. Apart from being induced by a genetic predisposition, ageing or viral infections (particularly cytomegalovirus infection), immunosenescence is accelerated in many inflammatory diseases and uraemia. This translates into an enhancement of vascular inflammation and cardiovascular disease varying from endothelial dysfunction to plaque rupture. Emerging data point to a mechanistic role of CD28null T cells in glomerulonephritis, where they initiate and propagate local inflammation in concordance with dendritic cells and macrophages. They are suitably equipped to escape immunological dampening by the absence of homing to lymph nodes, anti‐apoptotic properties and resistance to suppression by regulatory T cells. Early accumulation of senescent CD28null T cells precedes glomerular or vascular injury, and targeting these cells could open avenues for early treatment interventions that aim at abrogating a detrimental vicious cycle.
Collapse
Affiliation(s)
| | - Karel Van Damme
- Renal Division Ghent University Hospital Ghent Belgium
- Laboratory of Immunoregulation and Mucosal Immunology, Center for Inflammation Research VIB Center for Inflammation Research Ghent Belgium
| | | |
Collapse
|
2
|
Samstag Y, Bogert NV, Wabnitz GH, Din S, Therre M, Leuschner F, Katus HA, Konstandin MH. Reactive Oxidative Species-Modulated Ca 2+ Release Regulates β 2 Integrin Activation on CD4 + CD28 null T Cells of Acute Coronary Syndrome Patients. THE JOURNAL OF IMMUNOLOGY 2020; 205:2276-2286. [PMID: 32938726 DOI: 10.4049/jimmunol.2000327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 08/17/2020] [Indexed: 01/06/2023]
Abstract
The number and activity of T cell subsets in the atherosclerotic plaques are critical for the prognosis of patients with acute coronary syndrome. β2 Integrin activation is pivotal for T cell recruitment and correlates with future cardiac events. Despite this knowledge, differential regulation of adhesiveness in T cell subsets has not been explored yet. In this study, we show that in human T cells, SDF-1α-mediated β2 integrin activation is driven by a, so far, not-described reactive oxidative species (ROS)-regulated calcium influx. Furthermore, we show that CD4+CD28null T cells represent a highly reactive subset showing 25-fold stronger β2 integrin activation upon SDF-1α stimulation compared with CD28+ T cells. Interestingly, ROS-dependent Ca release was much more prevalent in the pathogenetically pivotal CD28null subset compared with the CD28+ T cells, whereas the established mediators of the classical pathways for β2 integrin activation (PKC, PI3K, and PLC) were similarly activated in both T cell subsets. Thus, interference with the calcium flux attenuates spontaneous adhesion of CD28null T cells from acute coronary syndrome patients, and calcium ionophores abolished the observed differences in the adhesion properties between CD28+ and CD28null T cells. Likewise, the adhesion of these T cell subsets was indistinguishable in the presence of exogenous ROS/H2O2 Together, these data provide a molecular explanation of the role of ROS in pathogenesis of plaque destabilization.
Collapse
Affiliation(s)
- Yvonne Samstag
- Institute of Immunology, Section Molecular Immunology, Ruprecht-Karls-University, D-69120 Heidelberg, Germany
| | - Nicolai V Bogert
- Department of Cardiology, Heidelberg University Hospital, Ruprecht-Karls-University, D-69120 Heidelberg, Germany; and.,German Centre for Cardiovascular Research Partner Site Heidelberg/Mannheim, Germany, Heidelberg University Hospital, D-69120 Heidelberg, Germany
| | - Guido H Wabnitz
- Institute of Immunology, Section Molecular Immunology, Ruprecht-Karls-University, D-69120 Heidelberg, Germany
| | - Shabana Din
- Department of Cardiology, Heidelberg University Hospital, Ruprecht-Karls-University, D-69120 Heidelberg, Germany; and.,German Centre for Cardiovascular Research Partner Site Heidelberg/Mannheim, Germany, Heidelberg University Hospital, D-69120 Heidelberg, Germany
| | - Markus Therre
- Department of Cardiology, Heidelberg University Hospital, Ruprecht-Karls-University, D-69120 Heidelberg, Germany; and.,German Centre for Cardiovascular Research Partner Site Heidelberg/Mannheim, Germany, Heidelberg University Hospital, D-69120 Heidelberg, Germany
| | - Florian Leuschner
- Department of Cardiology, Heidelberg University Hospital, Ruprecht-Karls-University, D-69120 Heidelberg, Germany; and.,German Centre for Cardiovascular Research Partner Site Heidelberg/Mannheim, Germany, Heidelberg University Hospital, D-69120 Heidelberg, Germany
| | - Hugo A Katus
- Department of Cardiology, Heidelberg University Hospital, Ruprecht-Karls-University, D-69120 Heidelberg, Germany; and.,German Centre for Cardiovascular Research Partner Site Heidelberg/Mannheim, Germany, Heidelberg University Hospital, D-69120 Heidelberg, Germany
| | - Mathias H Konstandin
- Department of Cardiology, Heidelberg University Hospital, Ruprecht-Karls-University, D-69120 Heidelberg, Germany; and .,German Centre for Cardiovascular Research Partner Site Heidelberg/Mannheim, Germany, Heidelberg University Hospital, D-69120 Heidelberg, Germany
| |
Collapse
|
3
|
Głogowska-Ligus J, Dąbek J. Expression profile of integrin family genes in patients with myocardial infarction and in healthy subjects: an oligonucleotide microarray and QRT-PCR assessment. Preliminary results. Minerva Med 2019; 110:224-231. [PMID: 30784245 DOI: 10.23736/s0026-4806.19.05842-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND Pathogenesis and clinical course of all known cardiovascular diseases are rooted in endothelial dysfunction. Coronary thrombosis which can aggravate the coronary condition leading to myocardial infarction (MI) is closely linked to cellular adhesion processes involving numerous adhesion molecules. The goal of our study was to find and quantitate the expression of integrin genes that differentiate between MI patients and healthy subjects. METHODS The study included 171 individuals, among whom 8 were chosen to provide study material for the oligonucleotide microarray investigation (4 MI patients and 4 healthy subjects). The investigated material consisted of RNA isolated from peripheral blood mononuclear cells. RESULTS Analysis of gene expression data from eight HG-U133A microarrays allowed identification of three genes differentiating the examined groups. The differentiating genes were found using the Bland-Altman method. Two of them showed increased expression (beta 2 integrin and beta 7 integrin genes), whereas expression level of the third (beta 3 integrin gene) was decreased. CONCLUSIONS The differences in integrin gene expression levels that were observed in MI patients, as compared to healthy individuals, might be responsible for endothelial dysfunction as well as rise in adhesion and aggregation processes in this group of patients and might lead to coronary vessel occlusion by thrombi and myocardial infarction.
Collapse
Affiliation(s)
- Joanna Głogowska-Ligus
- Department of Epidemiology, School of Public Health in Bytom, Medical University of Silesia in Katowice, Katowice, Poland -
| | - Józefa Dąbek
- Department of Cardiology, School of Health Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| |
Collapse
|
4
|
LaBute MX, Zhang X, Lenderman J, Bennion BJ, Wong SE, Lightstone FC. Adverse drug reaction prediction using scores produced by large-scale drug-protein target docking on high-performance computing machines. PLoS One 2014; 9:e106298. [PMID: 25191698 PMCID: PMC4156361 DOI: 10.1371/journal.pone.0106298] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 08/05/2014] [Indexed: 01/12/2023] Open
Abstract
Late-stage or post-market identification of adverse drug reactions (ADRs) is a significant public health issue and a source of major economic liability for drug development. Thus, reliable in silico screening of drug candidates for possible ADRs would be advantageous. In this work, we introduce a computational approach that predicts ADRs by combining the results of molecular docking and leverages known ADR information from DrugBank and SIDER. We employed a recently parallelized version of AutoDock Vina (VinaLC) to dock 906 small molecule drugs to a virtual panel of 409 DrugBank protein targets. L1-regularized logistic regression models were trained on the resulting docking scores of a 560 compound subset from the initial 906 compounds to predict 85 side effects, grouped into 10 ADR phenotype groups. Only 21% (87 out of 409) of the drug-protein binding features involve known targets of the drug subset, providing a significant probe of off-target effects. As a control, associations of this drug subset with the 555 annotated targets of these compounds, as reported in DrugBank, were used as features to train a separate group of models. The Vina off-target models and the DrugBank on-target models yielded comparable median area-under-the-receiver-operating-characteristic-curves (AUCs) during 10-fold cross-validation (0.60-0.69 and 0.61-0.74, respectively). Evidence was found in the PubMed literature to support several putative ADR-protein associations identified by our analysis. Among them, several associations between neoplasm-related ADRs and known tumor suppressor and tumor invasiveness marker proteins were found. A dual role for interstitial collagenase in both neoplasms and aneurysm formation was also identified. These associations all involve off-target proteins and could not have been found using available drug/on-target interaction data. This study illustrates a path forward to comprehensive ADR virtual screening that can potentially scale with increasing number of CPUs to tens of thousands of protein targets and millions of potential drug candidates.
Collapse
Affiliation(s)
- Montiago X LaBute
- Computational Engineering Division, Lawrence Livermore National Laboratory, Livermore, California, United States of America
| | - Xiaohua Zhang
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, United States of America
| | - Jason Lenderman
- Computational Engineering Division, Lawrence Livermore National Laboratory, Livermore, California, United States of America
| | - Brian J Bennion
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, United States of America
| | - Sergio E Wong
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, United States of America
| | - Felice C Lightstone
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, United States of America
| |
Collapse
|
5
|
Chaturvedi P, Tyagi SC. Epigenetic mechanisms underlying cardiac degeneration and regeneration. Int J Cardiol 2014; 173:1-11. [PMID: 24636549 DOI: 10.1016/j.ijcard.2014.02.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 12/29/2013] [Accepted: 02/08/2014] [Indexed: 01/08/2023]
Abstract
Epigenetic modifications which are defined by DNA methylation, histone modifications and microRNA mediated gene regulation, have been found to be associated with cardiac dysfunction and cardiac regeneration but the mechanisms are unclear. MicroRNA therapies have been proposed for cardiac regeneration and proliferation of stem cells into cardiomyocytes. Cardiovascular disorders are represented by abnormal methylation of CpG islands and drugs that inhibit DNA methyltransferases such as 5-methyl Aza cytidine are under trials. Histone modifications which include acetylation, methylation, phosphorylation, ADP ribosylation, sumoylation and biotinylation are represented within abnormal phenotypes of cardiac hypertrophy, cardiac development and contractility. MicroRNAs have been used efficiently to epigenetically reprogram fibroblasts into cardiomyocytes. MicroRNAs represent themselves as potential biomarkers for early detection of cardiac disorders which are difficult to diagnose and are captured at later stages. Because microRNAs regulate circadian genes, for example a nocturnin gene of circadian clockwork is regulated by miR122, they have a profound role in regulating biological clock and this may explain the high cardiovascular risk during the morning time. This review highlights the role of epigenetics which can be helpful in disease management strategies.
Collapse
Affiliation(s)
- Pankaj Chaturvedi
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, KY, USA
| | - Suresh C Tyagi
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, KY, USA.
| |
Collapse
|
6
|
Deddens JC, Colijn JM, Oerlemans MIFJ, Pasterkamp G, Chamuleau SA, Doevendans PA, Sluijter JPG. Circulating microRNAs as novel biomarkers for the early diagnosis of acute coronary syndrome. J Cardiovasc Transl Res 2013; 6:884-98. [PMID: 23897095 DOI: 10.1007/s12265-013-9493-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 06/18/2013] [Indexed: 12/18/2022]
Abstract
Small non-coding microRNAs (miRNAs) are important physiological regulators of post-transcriptional gene expression. miRNAs not only reside in the cytoplasm but are also stably present in several extracellular compartments, including the circulation. For that reason, miRNAs are proposed as diagnostic biomarkers for various diseases. Early diagnosis of acute coronary syndrome (ACS), especially non-ST elevated myocardial infarction and unstable angina pectoris, is essential for optimal treatment outcome, and due to the ongoing need for additional identifiers, miRNAs are of special interest as biomarkers for ACS. This review highlights the nature and cellular release mechanisms of circulating miRNAs and therefore their potential role in the diagnosis of myocardial infarction. We will give an update of clinical studies addressing the role of circulating miRNA expression after myocardial infarction and explore the diagnostic value of this potential biomarker.
Collapse
Affiliation(s)
- J C Deddens
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Heidelberglaan 100, room G02.523, 3584, CX, Utrecht, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
7
|
Holmes RS, Rout UK. Comparative studies of vertebrate Beta integrin genes and proteins: ancient genes in vertebrate evolution. Biomolecules 2011; 1:3-31. [PMID: 24970121 PMCID: PMC4030831 DOI: 10.3390/biom1010003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 08/14/2011] [Accepted: 08/15/2011] [Indexed: 12/31/2022] Open
Abstract
Intregins are heterodimeric α- and β-subunit containing membrane receptor proteins which serve various cell adhesion roles in tissue repair, hemostasis, immune response, embryogenesis and metastasis. At least 18 α- (ITA or ITGA) and 8 β-integrin subunits (ITB or ITGB) are encoded on mammalian genomes. Comparative ITB amino acid sequences and protein structures and ITB gene locations were examined using data from several vertebrate genome projects. Vertebrate ITB genes usually contained 13-16 coding exons and encoded protein subunits with ~800 amino acids, whereas vertebrate ITB4 genes contained 36-39 coding exons and encoded larger proteins with ~1800 amino acids. The ITB sequences exhibited several conserved domains including signal peptide, extracellular β-integrin, β-tail domain and integrin β-cytoplasmic domains. Sequence alignments of the integrin β-cytoplasmic domains revealed highly conserved regions possibly for performing essential functions and its maintenance during vertebrate evolution. With the exception of the human ITB8 sequence, the other ITB sequences shared a predicted 19 residue α-helix for this region. Potential sites for regulating human ITB gene expression were identified which included CpG islands, transcription factor binding sites and microRNA binding sites within the 3'-UTR of human ITB genes. Phylogenetic analyses examined the relationships of vertebrate beta-integrin genes which were consistent with four major groups: 1: ITB1, ITB2, ITB7; 2: ITB3, ITB5, ITB6; 3: ITB4; and 4: ITB8 and a common evolutionary origin from an ancestral gene, prior to the appearance of fish during vertebrate evolution. The phylogenetic analyses revealed that ITB4 is the most likely primordial form of the vertebrate β integrin subunit encoding genes, that is the only β subunit expressed as a constituent of the sole integrin receptor 'α6β4' in the hemidesmosomes of unicellular organisms.
Collapse
Affiliation(s)
- Roger S Holmes
- School of Biomolecular and Physical Sciences, Griffith University, Nathan, 4111QLD, Australia.
| | - Ujjwal K Rout
- Department of Surgery, University of Mississippi Medical Center, Jackson, MS 38677, USA.
| |
Collapse
|
8
|
Necrotic cell death in atherosclerosis. Basic Res Cardiol 2011; 106:749-60. [DOI: 10.1007/s00395-011-0192-x] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Revised: 05/10/2011] [Accepted: 05/11/2011] [Indexed: 02/06/2023]
|
9
|
Mitogen-activated protein kinases activation in T lymphocytes of patients with acute coronary syndromes. Basic Res Cardiol 2011; 106:667-79. [DOI: 10.1007/s00395-011-0172-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Revised: 02/17/2011] [Accepted: 03/09/2011] [Indexed: 10/18/2022]
|
10
|
Andrié RP, Bauriedel G, Braun P, Höpp HW, Nickenig G, Skowasch D. Prevalence of intimal heat shock protein 60 homologues in unstable angina and correlation with anti-heat shock protein antibody titers. Basic Res Cardiol 2011; 106:657-65. [PMID: 21416407 DOI: 10.1007/s00395-011-0171-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Revised: 02/16/2011] [Accepted: 03/04/2011] [Indexed: 12/21/2022]
Abstract
Heat shock proteins (HSPs) are among the most highly conserved and immunogenic proteins shared by microbial agents and mammals. Human (h) HSP60 is upregulated under stress conditions and serves as a target for cross-reactive cytotoxic HSP-serum-antibodies. The present study evaluates the expressions of hHSP60 and its homologue chlamydial (c) HSP60 in advanced human coronary lesions and correlates intimal tissue-bound HSP expressions with circulating HSP-antibodies. Coronary atherectomy specimens retrieved from 100 primary target lesions of patients with unstable angina (UA; n = 40) or stable angina (SA; n = 60) were assessed immunohistochemically for the presence of hHSP60 and cHSP60. In a subgroup (n = 40), blood samples were tested for anti-Chl. pn.-IgG/IgA-titers and anti-HSP65-antibody titers. Coronary plaques revealed immunoreactive hHSP60 in 55% and cHSP60 in 45% of the lesions. Expression of both HSP homologues was significantly (each p < 0.001) higher in UA lesions compared with SA lesions (7.4 vs. 1.2% and 6.0 vs. 1.1%). HSP homologues showed positive correlations both in UA- and SA-lesions (r = 0.41, 0.33; p < 0.05). cHSP60 showed no association with anti-Chl. pn.-IgG/IgA-titers, whereas expressions of both homologues correlated positive with anti-HSP65-Ab titers (r = 0.42, p < 0.05; r = 0.50, p < 0.01). Intimal amounts of HSP60 homologues were associated with increased expressions of C-reactive protein, Toll-like receptor-4 and tissue factor. Human and chlamydial HSP60 colocalize within coronary atheroma, most prevalent in lesions associated with UA. Our data demonstrate a significant correlation between the intimal expressions of HSP60 homologues and serum HSP65 antibodies, thereby suggesting that humoral immune reactions may play an important role in coronary atherosclerosis and plaque instability.
Collapse
Affiliation(s)
- René P Andrié
- Department of Internal Medicine II/Cardiology, University of Bonn, Sigmund-Freud-Strasse 25, Bonn, Germany.
| | | | | | | | | | | |
Collapse
|
11
|
Erbel C, Dengler TJ, Wangler S, Lasitschka F, Bea F, Wambsganss N, Hakimi M, Böckler D, Katus HA, Gleissner CA. Expression of IL-17A in human atherosclerotic lesions is associated with increased inflammation and plaque vulnerability. Basic Res Cardiol 2010; 106:125-34. [PMID: 21116822 DOI: 10.1007/s00395-010-0135-y] [Citation(s) in RCA: 140] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Revised: 10/25/2010] [Accepted: 11/09/2010] [Indexed: 01/11/2023]
Abstract
A chronic (auto)immune response is the critical mechanism in atherosclerosis. Interleukin-17A is a pivotal effector cytokine, which modulates immune cell trafficking and initiates inflammation in (auto)immune and infectious diseases. However, expression of IL-17A in the context of human atherosclerosis has hardly been explored. Carotid artery plaques were collected from 79 patients undergoing endarterectomy. Patients were grouped according to their symptomatic status (TIA, stroke), plaque morphology and medication. Quantitative RT-PCR was used to analyze tissue inflammation and immunohistochemistry to assess cellular source of IL-17A expression and lesion morphology. Carotid plaques from patients with ischemic symptoms were characterized by a highly activated inflammatory milieu including accumulation of T cells (p = 0.04) and expression of IL-6 and VCAM1 (p = 0.02, 0.01). Expression of IL-17A and its positive regulators IL-21 and IL-23 was present in atherosclerotic lesions, significantly upregulated in atheromas of symptomatic patients (p = 0.005, 0.004, 0.03), and expression of IL-17A and IL-21 showed a strong correlation (p = 0.002, r = 0.52). The cellular sources of lesional IL-17A expression are T cells, macrophages, B cells and plasma cells. Vulnerable/ruptured (complicated) plaques were significantly associated with IL-17A expression levels (p = 0.003). In addition, IL-17A showed a marked negative correlation with the potent anti-inflammatory/atheroprotective cytokine IL-10 (p = 0.0006, r = -0.46). Furthermore, treatment with a HMG-CoA reductase inhibitor or acetylsalicylic acid showed reduced levels of IL-21, IL-23 and VCAM1 (all p < 0.05), but did not influence IL-17A. The association of IL-17A with ischemic symptoms and vulnerable plaque characteristics suggests that the pro-inflammatory cytokine IL-17A may contribute to atherosclerosis und plaque instability.
Collapse
Affiliation(s)
- Christian Erbel
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Meder B, Keller A, Vogel B, Haas J, Sedaghat-Hamedani F, Kayvanpour E, Just S, Borries A, Rudloff J, Leidinger P, Meese E, Katus HA, Rottbauer W. MicroRNA signatures in total peripheral blood as novel biomarkers for acute myocardial infarction. Basic Res Cardiol 2010; 106:13-23. [PMID: 20886220 DOI: 10.1007/s00395-010-0123-2] [Citation(s) in RCA: 198] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Revised: 09/20/2010] [Accepted: 09/22/2010] [Indexed: 12/20/2022]
Abstract
MicroRNAs (miRNAs) are important regulators of adaptive and maladaptive responses in cardiovascular diseases and hence are considered to be potential therapeutical targets. However, their role as novel biomarkers for the diagnosis of cardiovascular diseases still needs to be systematically evaluated. We assessed here for the first time whole-genome miRNA expression in peripheral total blood samples of patients with acute myocardial infarction (AMI). We identified 121 miRNAs, which are significantly dysregulated in AMI patients in comparison to healthy controls. Among these, miR-1291 and miR-663b show the highest sensitivity and specificity for the discrimination of cases from controls. Using a novel self-learning pattern recognition algorithm, we identified a unique signature of 20 miRNAs that predicts AMI with even higher power (specificity 96%, sensitivity 90%, and accuracy 93%). In addition, we show that miR-30c and miR-145 levels correlate with infarct sizes estimated by Troponin T release. The here presented study shows that single miRNAs and especially miRNA signatures derived from peripheral blood, could be valuable novel biomarkers for cardiovascular diseases.
Collapse
Affiliation(s)
- Benjamin Meder
- Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|