1
|
Na SP, Ning ML, Ma JF, Liang S, Wang YL, Sui MS, Guo XF, Ji Y, Lyu HY, Yuan XY, Bao YS. Association of elevated circulating monocyte-platelet aggregates with hypercoagulability in patients with nephrotic syndrome. Thromb J 2024; 22:56. [PMID: 38943162 PMCID: PMC11212416 DOI: 10.1186/s12959-024-00626-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 06/19/2024] [Indexed: 07/01/2024] Open
Abstract
BACKGROUND Hypercoagulability emerges as a central pathological feature and clinical complication in nephrotic syndrome. Increased platelet activation and aggregability are closely related to hypercoagulability in nephrotic syndrome. Monocyte-platelet aggregates (MPAs) have been proposed to represent a robust biomarker of platelet activation. The aim of this study was to investigate levels of the circulating MPAs and MPAs with the different monocyte subsets to evaluate the association of MPAs with hypercoagulability in nephrotic syndrome. METHODS Thirty-two patients with nephrotic syndrome were enrolled. In addition, thirty-two healthy age and sex matched adult volunteers served as healthy controls. MPAs were identified by CD14 monocytes positive for CD41a platelets. The classical (CD14 + + CD16-, CM), the intermediate (CD14 + + CD16+, IM) and the non-classical (CD14 + CD16++, NCM) monocytes, as well as subset specific MPAs, were measured by flow cytometry. RESULTS Patients with nephrotic syndrome showed a higher percentage of circulating MPAs as compared with healthy controls (p < 0.001). The percentages of MPAs with CM, IM, and NCM were higher than those of healthy controls (p = 0.012, p < 0.001 and p < 0.001, respectively). Circulating MPAs showed correlations with hypoalbuminemia (r=-0.85; p < 0.001), hypercholesterolemia (r = 0.54; p < 0.001), fibrinogen (r = 0.70; p < 0.001) and D-dimer (r = 0.37; p = 0.003), but not with hypertriglyceridemia in nephrotic syndrome. The AUC for the prediction of hypercoagulability in nephrotic syndrome using MPAs was 0.79 (95% CI 0.68-0.90, p < 0.001). The sensitivity of MPAs in predicting hypercoagulability was 0.71, and the specificity was 0.78. CONCLUSION Increased MPAs were correlated with hypercoagulability in nephrotic syndrome. MPAs may serve as a potential biomarker for thrombophilic or hypercoagulable state and provide novel insight into the mechanisms of anticoagulation in nephrotic syndrome.
Collapse
Affiliation(s)
- Shi-Ping Na
- Department of Nephrology, First Affiliated Hospital of Harbin Medical University, Harbin Medical University, No.23 Youzheng Street, Harbin, 150001, China
| | - Mei-Liang Ning
- Department of Nephrology, First Affiliated Hospital of Harbin Medical University, Harbin Medical University, No.23 Youzheng Street, Harbin, 150001, China
| | - Ji-Fang Ma
- Department of Nephrology, First Affiliated Hospital of Harbin Medical University, Harbin Medical University, No.23 Youzheng Street, Harbin, 150001, China
| | - Shuang Liang
- Department of Nephrology, First Affiliated Hospital of Harbin Medical University, Harbin Medical University, No.23 Youzheng Street, Harbin, 150001, China
| | - Yan-Li Wang
- Department of Rheumatology, First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, 150001, China
| | - Man-Shu Sui
- Department of Nephrology, First Affiliated Hospital of Harbin Medical University, Harbin Medical University, No.23 Youzheng Street, Harbin, 150001, China
| | - Xiao-Fang Guo
- Department of Nephrology, First Affiliated Hospital of Harbin Medical University, Harbin Medical University, No.23 Youzheng Street, Harbin, 150001, China
| | - Ying Ji
- Department of Nephrology, First Affiliated Hospital of Harbin Medical University, Harbin Medical University, No.23 Youzheng Street, Harbin, 150001, China
| | - Hui-Yan Lyu
- Department of Nephrology, First Affiliated Hospital of Harbin Medical University, Harbin Medical University, No.23 Youzheng Street, Harbin, 150001, China
| | - Xue-Ying Yuan
- Department of Nephrology, First Affiliated Hospital of Harbin Medical University, Harbin Medical University, No.23 Youzheng Street, Harbin, 150001, China
| | - Yu-Shi Bao
- Department of Nephrology, First Affiliated Hospital of Harbin Medical University, Harbin Medical University, No.23 Youzheng Street, Harbin, 150001, China.
| |
Collapse
|
2
|
Zisman E, Hossain M, Funderburg NT, Christenson R, Jeudy J, Burrowes S, Hays AG, George N, Freeman ML, Rebuck H, Mitchell SE, Miller M, Bagchi S. Association of Lipoprotein(a) with peri-coronary inflammation in persons with and without HIV infection. J Clin Lipidol 2024; 18:e430-e443. [PMID: 38403541 PMCID: PMC11209819 DOI: 10.1016/j.jacl.2024.02.003] [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: 08/04/2023] [Revised: 12/21/2023] [Accepted: 02/09/2024] [Indexed: 02/27/2024]
Abstract
BACKGROUND Persons with human immunodeficiency virus (HIV) (PWH) have an increased risk of developing cardiovascular disease (CVD) compared to persons without HIV (PWoH). Lipoprotein(a) [Lp(a)] is a known atherosclerotic risk factor in PWoH, but there are no studies investigating Lp(a) and peri-coronary inflammation. OBJECTIVE To investigate whether Lp(a) is associated with peri-coronary inflammation as assessed by the fat attenuation index (FAI) and activated monocytes and T lymphocytes in PWH and PWoH. METHODS We measured plasma levels of Lp(a) at study entry in 58 PWH and 21 PWoH without CVD and who had FAI measurements. Associations of Lp(a) with FAI values of the right coronary artery (RCA) and left anterior descending artery were evaluated using multivariable regression models adjusted for potential confounders. Correlations between Lp(a) levels and systemic inflammatory markers and immune cell subsets were examined. RESULTS Lp(a) was associated with greater peri-coronary inflammation among PWH compared to PWoH (β=1.73, P=0.019) in the RCA, in adjusted models. Significant correlations were observed with certain inflammatory markers (tumor necrosis factor receptor [TNFR]-I, b=0.295, P<0.001; TNFR-II, b=0.270, P=0.002; high-sensitivity C-reactive protein, b=0.195, P=0.028). Significant correlations were found between Lp(a) levels and several markers of monocyte activation: CD16 -CD163+ (b= -0.199, P=0.024), and CD16 -DR+ MFI (b= -0.179, P=0.042) and T cell subset CD38+CD4+ TEMRA (b= 0.177, P= 0.044). CONCLUSIONS Lp(a) was associated with greater peri-coronary inflammation in the RCA in PWH compared to PWoH, as well as with select systemic inflammatory markers and specific subsets of immune cells in peripheral circulation.
Collapse
Affiliation(s)
- Erin Zisman
- University of Maryland School of Medicine, Department of Medicine, Baltimore, MD, USA (Dr Zisman)
| | - Mian Hossain
- Morgan State University, School of Community Health and Policy, Department of Statistics, Baltimore, MD, USA (Dr Hossain)
| | - Nicholas T Funderburg
- The Ohio State University, School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, Columbus, OH, USA (Dr Funderburg)
| | - Robert Christenson
- University of Maryland School of Medicine, Department of Pathology, Baltimore, MD, USA (Drs Christenson, Rebuck)
| | - Jean Jeudy
- University of Maryland School of Medicine, Department of Diagnostic Radiology and Nuclear Medicine, Baltimore, MD, USA (Dr Jeudy)
| | - Shana Burrowes
- Boston University School of Medicine, Department of Medicine, Section of Infectious Diseases, Boston, MA, USA (Dr Burrowes)
| | - Allison G Hays
- Johns Hopkins University, Department of Medicine, Division of Cardiology, Baltimore, MD, USA (Dr Hays)
| | - Nivya George
- University of Maryland School of Medicine, Institute of Human Virology (Drs George, Mitchell)
| | - Michael L Freeman
- Division of Infectious Diseases and HIV Medicine, Case Western Reserve University, Cleveland, OH, USA (Dr Freeman)
| | - Heather Rebuck
- University of Maryland School of Medicine, Department of Pathology, Baltimore, MD, USA (Drs Christenson, Rebuck)
| | - Sarah E Mitchell
- University of Maryland School of Medicine, Institute of Human Virology (Drs George, Mitchell)
| | - Michael Miller
- Corporal Michael J. Crescenz VA Medical Center & Hospital of the University of Pennsylvania, Department of Medicine, Philadelphia, PA, USA (Dr Miller)
| | - Shashwatee Bagchi
- Washington University in St. Louis, Division of Infectious Diseases, St. Louis, MO, USA (Dr Bagchi).
| |
Collapse
|
3
|
Merinopoulos I, Bhalraam U, Holmes T, Tsampasian V, Corballis N, Gunawardena T, Sawh C, Maart C, Wistow T, Ryding A, Eccleshall SC, Smith J, Vassiliou VS. Circulating intermediate monocytes CD14++CD16+ are increased after elective percutaneous coronary intervention. PLoS One 2023; 18:e0294746. [PMID: 38096193 PMCID: PMC10721025 DOI: 10.1371/journal.pone.0294746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 11/07/2023] [Indexed: 12/17/2023] Open
Abstract
AIM Inflammation plays a central role in the pathogenesis of atherosclerosis and in the sequelae of percutaneous coronary intervention (PCI). Previous work demonstrated that intermediate monocytes (CD14++CD16+) are associated with adverse cardiovascular events, yet monocyte subset response following elective PCI has not been described. This article explores the changes in monocyte subset and humoral response after elective PCI. METHODS This prospective study included 30 patients without inflammatory diseases being referred for elective PCI. We included patients treated with drug coated balloons or 2nd generation drug eluting stents. Patients underwent blood tests at baseline (prior to PCI), four hours, two weeks and two months later. Analyses were performed in terms of monocyte subsets (classical CD14++CD16-, intermediate CD14++CD16+ and non-classical CD14+CD16++), gene expression of CD14+ leucocytes and humoral biomarkers. RESULTS Intermediate monocytes decreased significantly four hours after PCI, were recovered at two weeks, and increased significantly at two months post elective, uncomplicated PCI. They remain significantly elevated in the DES group but not in the DCB group. Gene expression analysis of CD14+ leucocytes showed IL18 had decreased expression at two weeks, CXCR4 and IL1β decreased at two months, while pentraxin 3 increased at two weeks and two months. In terms of humoral biomarkers, hsTnI remains elevated up to two weeks post PCI while IL6 and TNFα remain elevated till two months post PCI. CONCLUSION Intermediate monocytes increase significantly two months following elective, uncomplicated PCI. They remain significantly elevated in the DES group but not in the DCB group suggesting that the PCI strategy could be one of the ways to modulate the inflammatory response post PCI.
Collapse
Affiliation(s)
- Ioannis Merinopoulos
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, United Kingdom
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - U Bhalraam
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, United Kingdom
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Terri Holmes
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Vasiliki Tsampasian
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, United Kingdom
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Natasha Corballis
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, United Kingdom
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Tharusha Gunawardena
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, United Kingdom
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Chris Sawh
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - Clint Maart
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - Trevor Wistow
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - Alisdair Ryding
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - Simon C. Eccleshall
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, United Kingdom
| | - James Smith
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Vassilios S. Vassiliou
- Department of Cardiology, Norfolk and Norwich University Hospital, Norwich, United Kingdom
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
- Institute of Continuing Education, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
4
|
Kologrivova IV, Suslova TE, Koshelskaya OA, Kravchenko ES, Kharitonova OA, Romanova EA, Vyrostkova AI, Boshchenko AA. Intermediate Monocytes and Circulating Endothelial Cells: Interplay with Severity of Atherosclerosis in Patients with Coronary Artery Disease and Type 2 Diabetes Mellitus. Biomedicines 2023; 11:2911. [PMID: 38001912 PMCID: PMC10669450 DOI: 10.3390/biomedicines11112911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
The aim was to investigate the association of monocyte heterogeneity and presence of circulating endothelial cells with the severity of coronary atherosclerosis in patients with coronary artery disease (CAD) and type 2 diabetes mellitus (T2DM). We recruited 62 patients with CAD, including 22 patients with DM2. The severity of atherosclerosis was evaluated using Gensini Score. Numbers of classical (CD14++CD16-), intermediate (CD14++CD16+), and non-classical (CD14+CD16++) monocyte subsets; circulating endothelial progenitor cells; and the presence of circulating endothelial cells were evaluated. Counts and frequencies of intermediate monocytes, but not glycaemia parameters, were associated with the severity of atherosclerosis in diabetic CAD patients (rs = 0.689; p = 0.001 and rs = 0.632; p = 0.002, respectively). Frequency of Tie2+ cells was lower in classical than in non-classical monocytes in CAD patients (p = 0.007), while in patients with association of CAD and T2DM, differences between Tie2+ monocytes subsets disappeared (p = 0.080). Circulating endothelial cells were determined in 100% of CAD+T2DM patients, and counts of CD14++CD16+ monocytes and concentration of TGF-β predicted the presence of circulating endothelial cells (sensitivity 92.3%; specificity 90.9%; AUC = 0.930). Thus, intermediate monocytes represent one of the key determinants of the appearance of circulating endothelial cells in all the patients with CAD, but are associated with the severity of atherosclerosis only in patients with association of CAD and T2DM.
Collapse
Affiliation(s)
- Irina V. Kologrivova
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111A Kievskaya, Tomsk 634012, Russia; (T.E.S.); (O.A.K.); (E.S.K.); (O.A.K.); (A.A.B.)
| | - Tatiana E. Suslova
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111A Kievskaya, Tomsk 634012, Russia; (T.E.S.); (O.A.K.); (E.S.K.); (O.A.K.); (A.A.B.)
| | - Olga A. Koshelskaya
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111A Kievskaya, Tomsk 634012, Russia; (T.E.S.); (O.A.K.); (E.S.K.); (O.A.K.); (A.A.B.)
| | - Elena S. Kravchenko
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111A Kievskaya, Tomsk 634012, Russia; (T.E.S.); (O.A.K.); (E.S.K.); (O.A.K.); (A.A.B.)
| | - Olga A. Kharitonova
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111A Kievskaya, Tomsk 634012, Russia; (T.E.S.); (O.A.K.); (E.S.K.); (O.A.K.); (A.A.B.)
| | - Ekaterina A. Romanova
- Department of Biomedicine, Siberian State Medical University, 2 Moskovskii trakt, Tomsk 634050, Russia; (E.A.R.); (A.I.V.)
| | - Alexandra I. Vyrostkova
- Department of Biomedicine, Siberian State Medical University, 2 Moskovskii trakt, Tomsk 634050, Russia; (E.A.R.); (A.I.V.)
| | - Alla A. Boshchenko
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111A Kievskaya, Tomsk 634012, Russia; (T.E.S.); (O.A.K.); (E.S.K.); (O.A.K.); (A.A.B.)
| |
Collapse
|
5
|
Ruder AV, Wetzels SMW, Temmerman L, Biessen EAL, Goossens P. Monocyte heterogeneity in cardiovascular disease. Cardiovasc Res 2023; 119:2033-2045. [PMID: 37161473 PMCID: PMC10478755 DOI: 10.1093/cvr/cvad069] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/07/2023] [Accepted: 02/21/2023] [Indexed: 05/11/2023] Open
Abstract
Monocytes circulate the vasculature at steady state and are recruited to sites of inflammation where they differentiate into macrophages (MФ) to replenish tissue-resident MФ populations and engage in the development of cardiovascular disease (CVD). Monocytes display considerable heterogeneity, currently reflected by a nomenclature based on their expression of cluster of differentiation (CD) 14 and CD16, distinguishing CD14++CD16- classical (cMo), CD14++CD16+ intermediate (intMo) and CD14+CD16++ non-classical (ncMo) monocytes. Several reports point to shifted subset distributions in the context of CVD, with significant association of intMo numbers with atherosclerosis, myocardial infarction, and heart failure. However, clear indications of their causal involvement as well as their predictive value for CVD are lacking. As recent high-parameter cytometry and single-cell RNA sequencing (scRNA-Seq) studies suggest an even higher degree of heterogeneity, better understanding of the functionalities of these subsets is pivotal. Considering their high heterogeneity, surprisingly little is known about functional differences between MФ originating from monocytes belonging to different subsets, and implications thereof for CVD pathogenesis. This paper provides an overview of recent findings on monocyte heterogeneity in the context of homeostasis and disease as well as functional differences between the subsets and their potential to differentiate into MФ, focusing on their role in vessels and the heart. The emerging paradigm of monocyte heterogeneity transcending the current tripartite subset division argues for an updated nomenclature and functional studies to substantiate marker-based subdivision and to clarify subset-specific implications for CVD.
Collapse
Affiliation(s)
- Adele V Ruder
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC+), P. Debyelaan 25, 6229 HX Maastricht, The Netherlands
| | - Suzan M W Wetzels
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC+), P. Debyelaan 25, 6229 HX Maastricht, The Netherlands
| | - Lieve Temmerman
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC+), P. Debyelaan 25, 6229 HX Maastricht, The Netherlands
| | - Erik A L Biessen
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC+), P. Debyelaan 25, 6229 HX Maastricht, The Netherlands
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Pieter Goossens
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC+), P. Debyelaan 25, 6229 HX Maastricht, The Netherlands
| |
Collapse
|
6
|
Filatova AY, Afanasieva OI, Arefieva TI, Potekhina AV, Tyurina AV, Klesareva EA, Razova OA, Ezhov MV, Pokrovsky SN. The Concentration of PCSK9-Lp(a) Complexes and the Level of Blood Monocytes in Males with Coronary Atherosclerosis. J Pers Med 2023; 13:1077. [PMID: 37511689 PMCID: PMC10381556 DOI: 10.3390/jpm13071077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
In this study we analyzed the concentration of lipoprotein(a) (Lp(a)), PCSK9-Lp(a) complexes and the circulating monocyte subsets in coronary atherosclerosis. For this study, 257 patients with coronary atherosclerosis and 68 patients without stenotic atherosclerosis in the coronary, carotid and lower extremity arteries (control group) were enrolled. The monocyte subpopulations (classical CD14++CD16-, intermediate CD14++CD16+ and non-classical CD14+CD16++) were analyzed by direct immunofluorescence and flow cytometry. The Lp(a) and PCSK9-Lp(a) complexes in the serum were detected by ELISA. The concentration of Lp(a) was higher in the coronary atherosclerosis group compared with the controls (23.0 (9.1; 73.3) mg/dL versus 10.7 (4.7; 25.0) mg/dL, p < 0.05). No correlations between the level of Lp(a) and the concentration of the PCSK9-Lp(a) complexes, nor between the level of Lp(a) or PCSK9 and the total number of monocytes, were observed in either group. A slight positive correlation between the concentration of PCSK9-Lp(a) complexes and the absolute level of monocytes was obtained (r = 0.20, p = 0.002) in the patients with atherosclerosis due to the intermediate monocyte subsets (r = 0.33, p = 0.04). According to regression analysis, both the PCSK9-Lp(a) complexes concentration and BMI were related to the absolute number of blood monocytes in patients with atherosclerosis. Further studies are required to determine the pathogenetic contribution of PCSK9-Lp(a) complexes to the development of atherosclerosis.
Collapse
Affiliation(s)
- Anastasiia Yu Filatova
- Institute of Experimental Cardiology, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Olga I Afanasieva
- Institute of Experimental Cardiology, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Tatiana I Arefieva
- Institute of Experimental Cardiology, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Alexandra V Potekhina
- A.L. Myasnikov Institute of Clinical Cardiology, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Alexandra V Tyurina
- A.L. Myasnikov Institute of Clinical Cardiology, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Elena A Klesareva
- Institute of Experimental Cardiology, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Oksana A Razova
- Institute of Experimental Cardiology, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Marat V Ezhov
- A.L. Myasnikov Institute of Clinical Cardiology, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Sergey N Pokrovsky
- Institute of Experimental Cardiology, National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| |
Collapse
|
7
|
Mendoza MF, Lavie CJ. Clinical associations between exercise and lipoproteins. Curr Opin Lipidol 2022; 33:364-373. [PMID: 36305382 DOI: 10.1097/mol.0000000000000848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
PURPOSE OF REVIEW One of the major concerns in global health is the deteriorating control of dyslipidemia (DLD), which is a very strong modifiable risk factor for untoward cardiovascular disease (CVD) outcomes. It serves as a foundation for atherosclerotic lesions that can be destabilized by co-inflammatory processes leading to distal clot migration and other related CVD events. There are many misconceptions regarding the management of DLD. Many health sectors advocate for weight loss without a clear-cut target to achieve better CVD outcomes. There is growing evidence that exercise training compliance regardless of weight change is a more reliable indicator of favorable outcomes. This review is intended to understand the relationship between exercise training, lipoprotein readings, and with CVD and all-cause morbidity and mortality. RECENT FINDINGS Aerobic exercise training (aET) and resistance exercise training (rET) increase cardiorespiratory fitness (CRF) and muscular strength (MusS), respectively. Regardless of weight loss, aET and rET are both known to independently reduce mortality possibly partly through improvement of lipid profiles. Of the two modes of exercise, rET has propensity for enhanced compliance because of its significant lipid and mortality-attenuating effect even with just brief exercise sessions. However, there are several studies showing that participation in both modes of exercise causes more pronounced improvements in DLD and CVD-related mortality compared with either mode of exercise training alone. In addition, Lipoprotein-a [Lp(a)] has been increasingly acknowledged to be atherogenic because of its LDL core. The close proximity of Lp(a) with macrophages triggers the development of atheromas, plaque formation, and growth. This causes a cascade of inflammatory processes that increase the development of ischemic CVD and calcific aortic valve stenosis. Although exercise training is known to reduce plasma LDL-C levels, it has no direct effect on Lp(a) levels as the latter lipoprotein is not influenced by motion nor exercise. Reviews of multiple studies lead us to infer that exercise training may potentially have an indirect impact on Lp(a) attenuation because of the ability of exercise training to inhibit Proprotein Convertase Subtisilin/Kexin type-9 (PCSK-9), as some studies using pharmacologic therapy with PCSK-9 inhibitors were able to show a concomitant decrease in Lp(a) levels. SUMMARY It is clear that normal-to-overweight populations who are highly active have better CVD outcomes and lipid profiles than their sedentary counterparts, and those who were underweight and unfit fared much worse. This allows us to take a more precise approach in the management of DLD rather than plainly focusing on gross weight in patients. Exercise training certainly has beneficial impact on longevity owing to its advantageous effect on lipoprotein levels and particle size. As such, reputable health societies, such as the ESC, ACC, and AHA have prescribed the ideal exercise training regimen, which have noticeable similarities. Increasing the use of wearable devices may help improve our ability to prescribe, quantify, and precisely track physical activity in our continuing efforts to combat increasing morbidity related to unhealthy lifestyles and inactivity.
Collapse
Affiliation(s)
- Michael F Mendoza
- Department of Cardiovascular Diseases, John Ochsner Heart and Vascular Institute, Ochsner Clinical School, The University of Queensland School of Medicine
- The Gayle and Tom Benson Cancer Center, New Orleans, Louisiana, USA
| | - Carl J Lavie
- Department of Cardiovascular Diseases, John Ochsner Heart and Vascular Institute, Ochsner Clinical School, The University of Queensland School of Medicine
| |
Collapse
|
8
|
Abstract
Cardiovascular diseases (CVD) remain the leading cause of death globally, and further efforts are being undertaken to understand and modify CVD risk factors, such as dyslipidemia (DLD), hypertension, and diabetes. The sedentary lifestyle of most individuals today contributes to the prevalence of these conditions. Uncontrolled dyslipidemia serves as a fertile ground for atherosclerotic plaque formation, while lipoproteins (Lp) act as cofactors for inflammatory processes that cause plaque destabilization leading to subsequent CVD events. As such, many health experts and institutions continue to emphasize the importance of cardiorespiratory fitness (CRF) and muscular strength (MusS) with the intent to reduce atherogenic lipoproteins and proprotein convertase subtilisin kexin type 9 (PCSK-9) expression. Concordantly, the two modes of exercise training (ET), such as aerobic ET (aET) and resistance ET (rET) have both demonstrated to improve CRF and MusS, respectively. Although both modes of ET were shown to independently reduce mortality, participation in both forms resulted in a more pronounced improvement in cholesterol levels and CVD-related mortality. Though reduction of adiposity is not a pre-requisite to achieve better control of DLD through increased CRF and MusS, the beneficial effects of physical activity on the inflammatory processes linked to atherosclerosis are almost always associated with a simultaneous decrease in overall adiposity. It is therefore essential to promote both aET and rET, including weight loss in order to attenuate the risks stemming from atherosclerosis and its proinflammatory components.
Collapse
|
9
|
Innate immune cells in the pathophysiology of calcific aortic valve disease: lessons to be learned from atherosclerotic cardiovascular disease? Basic Res Cardiol 2022; 117:28. [PMID: 35581364 PMCID: PMC9114076 DOI: 10.1007/s00395-022-00935-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/04/2022] [Accepted: 05/04/2022] [Indexed: 01/31/2023]
Abstract
Calcific aortic valve disease (CAVD) is the most common valvular disease in the developed world with currently no effective pharmacological treatment available. CAVD results from a complex, multifactorial process, in which valvular inflammation and fibro-calcific remodelling lead to valve thickening and cardiac outflow obstruction. The exact underlying pathophysiology of CAVD is still not fully understood, yet the development of CAVD shows many similarities with the pathophysiology of atherosclerotic cardiovascular disease (ASCVD), such as coronary artery disease. Innate immune cells play a crucial role in ASCVD and might also play a pivotal role in the development of CAVD. This review summarizes the current knowledge on the role of innate immune cells, both in the circulation and in the aortic valve, in the development of CAVD and the similarities and differences with ASCVD. Trained immunity and clonal haematopoiesis of indeterminate potential are proposed as novel immunological mechanisms that possibly contribute to the pathophysiology of CAVD and new possible treatment targets are discussed.
Collapse
|
10
|
Abstract
The World Health Organization has declared obesity to be a global epidemic that increases cardiovascular disease (CVD) mortality risk factors, such as hypertension, diabetes, dyslipidemia, and atherosclerosis. The increasing ratio of time spent in sedentary activities to that spent performing physically demanding tasks increases the trends to obesity and susceptibility to these risk factors. Dyslipidemia is the foundation of atherosclerotic buildup and lipoproteins serve as cofactors to the inflammatory processes that destabilize plaques. Increasing cardiorespiratory fitness and muscular strength helps attenuate concentrations of low-density lipoproteins (LDLs), such as LDL cholesterol, and increase levels of high-density lipoprotein cholesterol, as well as reduce proprotein convertase subtilisin kexin type 9 expression. Effects of physical activity on the inflammatory pathways of atherosclerosis, specifically C-reactive protein, are more closely related to reducing the levels of adiposity in tandem with increasing fitness, than with exercise training alone. The purpose of this review is to describe the physiology of dyslipidemia and relate it to CVD and exercise therapies.
Collapse
|
11
|
Williams H, Mack CD, Li SCH, Fletcher JP, Medbury HJ. Nature versus Number: Monocytes in Cardiovascular Disease. Int J Mol Sci 2021; 22:ijms22179119. [PMID: 34502027 PMCID: PMC8430468 DOI: 10.3390/ijms22179119] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 01/01/2023] Open
Abstract
Monocytes play a key role in cardiovascular disease (CVD) as their influx into the vessel wall is necessary for the development of an atherosclerotic plaque. Monocytes are, however, heterogeneous differentiating from classical monocytes through the intermediate subset to the nonclassical subset. While it is recognized that the percentage of intermediate and nonclassical monocytes are higher in individuals with CVD, accompanying changes in inflammatory markers suggest a functional impact on disease development that goes beyond the increased proportion of these ‘inflammatory’ monocyte subsets. Furthermore, emerging evidence indicates that changes in monocyte proportion and function arise in dyslipidemia, with lipid lowering medication having some effect on reversing these changes. This review explores the nature and number of monocyte subsets in CVD addressing what they are, when they arise, the effect of lipid lowering treatment, and the possible implications for plaque development. Understanding these associations will deepen our understanding of the clinical significance of monocytes in CVD.
Collapse
Affiliation(s)
- Helen Williams
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, Sydney, NSW 2145, Australia; (H.W.); (C.D.M.); (J.P.F.)
- Westmead Clinical School, The University of Sydney, Westmead, Sydney, NSW 2145, Australia
| | - Corinne D. Mack
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, Sydney, NSW 2145, Australia; (H.W.); (C.D.M.); (J.P.F.)
- Westmead Clinical School, The University of Sydney, Westmead, Sydney, NSW 2145, Australia
| | - Stephen C. H. Li
- Chemical Pathology, NSW Health Pathology, Westmead Hospital and Institute of Clinical Pathology and Medical Research, Westmead, Sydney, NSW 2145, Australia;
- Blacktown/Mt Druitt Clinical School, Blacktown Hospital, Western Sydney University, Blacktown, NSW 2148, Australia
| | - John P. Fletcher
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, Sydney, NSW 2145, Australia; (H.W.); (C.D.M.); (J.P.F.)
- Westmead Clinical School, The University of Sydney, Westmead, Sydney, NSW 2145, Australia
| | - Heather J. Medbury
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, Sydney, NSW 2145, Australia; (H.W.); (C.D.M.); (J.P.F.)
- Westmead Clinical School, The University of Sydney, Westmead, Sydney, NSW 2145, Australia
- Correspondence:
| |
Collapse
|
12
|
Fiorelli S, Anesi A, Porro B, Cosentino N, Werba JP, Di Minno A, Manega CM, Barbieri S, Colombo GI, Marenzi G, Cavalca V, Tremoli E, Eligini S. Lipidomics analysis of monocytes from patients with acute myocardial infarction reveals lactosylceramide as a new player in monocyte migration. FASEB J 2021; 35:e21494. [PMID: 33856696 DOI: 10.1096/fj.202001872rrr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 02/15/2021] [Accepted: 02/17/2021] [Indexed: 11/11/2022]
Abstract
Monocyte recruitment after vascular injury and their migration through the vessel wall represent crucial events in the initiation, progression, and destabilization of atherosclerotic plaque. Circulating monocytes are exposed to stimuli that alter their physiological state, and among them, lipids play a key role. Several studies investigated the mechanisms by which lipids affect monocyte functions promoting coronary atherosclerotic plaque initiation, but information on the relationship between lipid composition and function of monocyte is scant. We aimed at studying the migration of circulating monocytes isolated from patients with acute myocardial infarction (AMI) at hospital presentation and investigating its correlation with cellular lipid profile. The migration of monocytes was tested using both fetal bovine serum (FBS) and autologous serum as chemoattractant stimuli. Monocyte lipid profile was evaluated through an untargeted lipidomics approach, using a liquid chromatography/time-of-flight mass spectrometry platform. We observed that AMI patients' monocytes showed a significant increase in FBS and autologous serum-mediated migration compared to controls. Moreover, a different monocyte lipidomic profile between the two study groups was detected. In particular, AMI patients' monocytes showed an altered composition in ceramides, with an increase in lactosylceramide and in phospholipids (ie, phosphatidylethanolamine and lisophosphatidylethanolamine). Of note, a positive correlation between lactosylceramide levels and monocyte migration was observed. Furthermore, the lactosylceramide synthase inhibition significantly reduced FBS-induced monocyte migration. Our results highlight the influence of lactosylceramide on the monocyte migration capacity, pointing out a new possible mechanism of lipids in the onset of atherothrombosis and, hence, in AMI.
Collapse
Affiliation(s)
| | - Andrea Anesi
- Centro Cardiologico Monzino I.R.C.C.S, Milan, Italy
| | | | | | - José P Werba
- Centro Cardiologico Monzino I.R.C.C.S, Milan, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
The Association of Lipoprotein(a) and Circulating Monocyte Subsets with Severe Coronary Atherosclerosis. J Cardiovasc Dev Dis 2021; 8:jcdd8060063. [PMID: 34206012 PMCID: PMC8228191 DOI: 10.3390/jcdd8060063] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/21/2021] [Accepted: 05/25/2021] [Indexed: 01/02/2023] Open
Abstract
Background and aims: Chronic inflammation associated with the uncontrolled activation of innate and acquired immunity plays a fundamental role in all stages of atherogenesis. Monocytes are a heterogeneous population and each subset contributes differently to the inflammatory process. A high level of lipoprotein(a) (Lp(a)) is a proven cardiovascular risk factor. The aim of the study was to investigate the association between the increased concentration of Lp(a) and monocyte subpopulations in patients with a different severity of coronary atherosclerosis. Methods: 150 patients (124 males) with a median age of 60 years undergoing a coronary angiography were enrolled. Lipids, Lp(a), autoantibodies, blood cell counts and monocyte subpopulations (classical, intermediate, non-classical) were analyzed. Results: The patients were divided into two groups depending on the Lp(a) concentration: normal Lp(a) < 30 mg/dL (n = 82) and hyperLp(a) ≥ 30 mg/dL (n = 68). Patients of both groups were comparable by risk factors, autoantibody levels and blood cell counts. In patients with hyperlipoproteinemia(a) the content (absolute and relative) of non-classical monocytes was higher (71.0 (56.6; 105.7) vs. 62.2 (45.7; 82.4) 103/mL and 17.7 (13.0; 23.3) vs. 15.1 (11.4; 19.4) %, respectively, p < 0.05). The association of the relative content of non-classical monocytes with the Lp(a) concentration retained a statistical significance when adjusted for gender and age (r = 0.18, p = 0.03). The severity of coronary atherosclerosis was associated with the Lp(a) concentration as well as the relative and absolute (p < 0.05) content of classical monocytes. The high content of non-classical monocytes (OR = 3.5, 95% CI 1.2–10.8) as well as intermediate monocytes (OR = 8.7, 2.5–30.6) in patients with hyperlipoproteinemia(a) were associated with triple-vessel coronary disease compared with patients with a normal Lp(a) level and a low content of monocytes. Conclusion: Hyperlipoproteinemia(a) and a decreased quantity of classical monocytes were associated with the severity of coronary atherosclerosis. The expansion of CD16+ monocytes (intermediate and non-classical) in the presence of hyperlipoproteinemia(a) significantly increased the risk of triple-vessel coronary disease.
Collapse
|
14
|
Cybularz M, Wydra S, Berndt K, Poitz DM, Barthel P, Alkouri A, Heidrich FM, Ibrahim K, Jellinghaus S, Speiser U, Linke A, Christoph M, Pfluecke C. Frailty is associated with chronic inflammation and pro-inflammatory monocyte subpopulations. Exp Gerontol 2021; 149:111317. [PMID: 33744391 DOI: 10.1016/j.exger.2021.111317] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 01/19/2023]
Abstract
AIM OF THE STUDY Frail patients with high grade aortic valve stenosis (AS) undergoing Transcatheter Aortic Valve Implantation (TAVI) have an increased mortality. A connection between frailty and inflammation has been suggested. Monocyte subpopulations are associated with both cardiovascular diseases and chronic inflammatory diseases. This study investigates the association of frailty with monocyte subpopulations and systemic inflammatory parameters in elderly patients undergoing TAVI. METHODS A total of 120 patients with symptomatic AS was examined. Before TAVI implantation, frailty was assessed by a bedside evaluation (eyeball test). In all patients a flow cytometry analysis has been performed. Monocyte subpopulations were defined as follows: classical (CD14++CD16-), intermediate (CD14++CD16+) and non-classical (CD14+CD16++). Expression of CD11b was measured as a marker for monocyte activation. Pro-inflammatory cytokines such as interleukin IL-8, as well as CRP were measured with Cytometric Bead Array or standard laboratory methods. RESULTS 28 out of 120 patients were frail. These patients showed both, signs of elevated chronic systemic inflammation reflected by elevated CRP (3.7 (1.4-5.4) vs. 5.9 (3.7-29.1), p = 0.001) and an elevated level of intermediate monocytes (37 (24-54) vs. 53 (47-63), p = 0.001). At 6 months after TAVI, 19 of 120 patients died, primarily without relevant dysfunction of the implanted aortic valve. Mortality was significantly higher in the frail as compared with non-frail patients (9 of 28 frail patients vs. 10 of 92 non frail patients, p < 0.001). A binary logistic regression analysis validated frailty and intermediate monocytes as independent predictors for early mortality after TAVI. CONCLUSION Chronic systemic inflammation and increased levels of intermediate monocytes are associated with frailty in old patients with severe aortic valve stenosis. Both the syndrome of frailty and elevated intermediate monocytes showed an association with early mortality after TAVI.
Collapse
Affiliation(s)
- Maria Cybularz
- Internal medicine, cardiology and intensive care medicine, Heart Center Dresden, Technical University Dresden, Dresden, Germany
| | - Sandy Wydra
- Internal medicine, cardiology and intensive care medicine, Heart Center Dresden, Technical University Dresden, Dresden, Germany
| | - Katharina Berndt
- Department of Cardiology, Universitätsklinikum Leipzig, Leipzig, Germany
| | - David M Poitz
- Institute for Clinical Chemistry and Laboratory Medicine, Technical University Dresden, Dresden, Germany
| | - Peggy Barthel
- Internal medicine, cardiology and intensive care medicine, Heart Center Dresden, Technical University Dresden, Dresden, Germany
| | - Ahmad Alkouri
- Internal medicine, cardiology and intensive care medicine, Heart Center Dresden, Technical University Dresden, Dresden, Germany
| | - Felix M Heidrich
- Internal medicine, cardiology and intensive care medicine, Heart Center Dresden, Technical University Dresden, Dresden, Germany
| | - Karim Ibrahim
- Department of Cardiology, Technische Universität Dresden, Klinikum Chemnitz, Germany
| | - Stefanie Jellinghaus
- Internal medicine, cardiology and intensive care medicine, Heart Center Dresden, Technical University Dresden, Dresden, Germany
| | - Uwe Speiser
- Internal medicine, cardiology and intensive care medicine, Heart Center Dresden, Technical University Dresden, Dresden, Germany
| | - Axel Linke
- Internal medicine, cardiology and intensive care medicine, Heart Center Dresden, Technical University Dresden, Dresden, Germany
| | - Marian Christoph
- Internal medicine, cardiology and intensive care medicine, Heart Center Dresden, Technical University Dresden, Dresden, Germany
| | - Christian Pfluecke
- Internal medicine, cardiology and intensive care medicine, Heart Center Dresden, Technical University Dresden, Dresden, Germany.
| |
Collapse
|
15
|
Coassin S, Kronenberg F. Mechanistic insights into lipoprotein(a): from infamous to ‘inflammous’. Eur Heart J 2020; 41:2272-2274. [DOI: 10.1093/eurheartj/ehaa420] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Stefan Coassin
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Kronenberg
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
16
|
Gorabi AM, Penson PE, Banach M, Motallebnezhad M, Jamialahmadi T, Sahebkar A. Epigenetic control of atherosclerosis via DNA methylation: A new therapeutic target? Life Sci 2020; 253:117682. [PMID: 32387418 DOI: 10.1016/j.lfs.2020.117682] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 04/01/2020] [Accepted: 04/15/2020] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is a disease in which lipid-laden plaques are developed inside the vessel walls of arteries. The immune system is activated, resulting in inflammation and oxidative stress. Endothelial cells (ECs) are activated, arterial smooth muscle cells (SMCs) proliferate, macrophages are activated, and foam cells are developed, leading to dysfunctional ECs. Epigenetic regulatory mechanisms, including DNA methylation, histone modifications, and microRNAs are involved in the modulation of genes that play distinct roles in several aspects of cell biology and physiology, hence linking environmental stimuli to gene regulation. Recent research has investigated the involvement of DNA methylation in the etiopathogenesis of atherosclerosis, and several studies have documented the role of this mechanism in various aspects of the disease. Regulation of DNA methylation plays a critical role in the integrity of ECs, SMC proliferation and formation of atherosclerotic lesions. In this review, we seek to clarify the role of DNA methylation in the development of atherosclerosis through different mechanisms.
Collapse
Affiliation(s)
- Armita Mahdavi Gorabi
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Peter E Penson
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Zeromskiego 113, Lodz, Poland; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| | - Morteza Motallebnezhad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Tannaz Jamialahmadi
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
17
|
Haybar H, Maleki Behzad M, Shahrabi S, Ansari N, Saki N. Expression of Blood Cells Associated CD Markers and Cardiovascular Diseases: Clinical Applications in Prognosis. Lab Med 2020; 51:122-142. [PMID: 31340048 DOI: 10.1093/labmed/lmz049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Cardiovascular diseases (CVDs) are a major cause of mortality worldwide. The results of various studies have shown that abnormality in the frequency and function of blood cells can be involved in CVD complications. In this review, we have focused on abnormalities in the expression of the CD (cluster of differentiation) markers of blood cells to assess the association of these abnormalities with CVD prognosis. METHODS We identified the relevant literature through a PubMed search (1990-2018) of English-language articles using the terms "Cardiovascular diseases", "CD markers", "leukocytes", "platelets", and "endothelial cells". RESULTS There is a variety of mechanisms for the effect of CD-marker expressions on CVDs prognosis, ranging from proinflammatory processes to dysfunctional effects in blood cells. CONCLUSION Considering the possible effects of CD-marker expression on CVDs prognosis, particularly prognosis of acute myocardial infarction and atherosclerosis, long-term studies in large cohorts are required to identify the prognostic value of CD markers and to target them with appropriate therapeutic agents.
Collapse
Affiliation(s)
- Habib Haybar
- Atherosclerosis Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Masumeh Maleki Behzad
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Saeid Shahrabi
- Department of Biochemistry and Hematology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Narges Ansari
- Isfahan Bone Metabolic Disorders Research Center, Department of Internal Medicine, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Najmaldin Saki
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
18
|
Figueroa-Vega N, Marín-Aragón CI, López-Aguilar I, Ibarra-Reynoso L, Pérez-Luque E, Malacara JM. Analysis of the percentages of monocyte subsets and ILC2s, their relationships with metabolic variables and response to hypocaloric restriction in obesity. PLoS One 2020; 15:e0228637. [PMID: 32074122 PMCID: PMC7029876 DOI: 10.1371/journal.pone.0228637] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 01/21/2020] [Indexed: 01/17/2023] Open
Abstract
PURPOSE Obesity results from excess energy intake over expenditure and is characterized by chronic low-grade inflammation involving circulating monocytes (Mo) and group 2 innate lymphoid cells (ILC2s) imbalance. We analyzed circulating Mo subsets and ILC2s percentages and β2-adrenergic receptor (β2AR) expression in lean and obese subjects, and the possible effect of hypocaloric restriction on these innate immune cells. METHODS In 139 individuals aged 45 to 57 years, classified in 74 lean individuals (>18.9kg/m2 BMI <24.9kg/m2) and 65 with obesity (n = 65), we collected fasting blood samples to detect Mo subsets, ILC2s number, and β2AR expression by flow cytometry. Lipids, insulin, leptin, and acylated-ghrelin concentrations were quantified. Resting energy expenditure (REE) was estimated by indirect calorimetry. These measurements were repeated in obese subjects after 7-weeks of hypocaloric restriction. RESULTS Non-classical monocytes (NCM) and β2AR expression on intermediate Mo (IM) were increased in obese individuals (p<0.001, in both cases), whereas the percent of ILC2s was decreased (p<0.0001). Stepwise regression analysis showed significantly negative associations of ILC2s with caloric intake, β2AR expression on IM with REE, but a positive relationship between NCM and HOMA-IR. Caloric restriction allowed a significant diminution of NCM and the β2AR expression on IM, as well as, an increase in the percent of classical Mo (CM), and ILC2s. ΔREE was related to ΔCD16+/CD16- ratio. CONCLUSIONS These findings show that in obesity occur changes in NCM, ILC2s and β2AR expression, which contribute to the low-grade inflammation linked to obesity and might revert with caloric restriction.
Collapse
Affiliation(s)
- Nicté Figueroa-Vega
- Department of Medical Sciences, University of Guanajuato, León Campus, León, Gto., México
| | | | - Itzel López-Aguilar
- Department of Medical Sciences, University of Guanajuato, León Campus, León, Gto., México
| | - Lorena Ibarra-Reynoso
- Department of Medical Sciences, University of Guanajuato, León Campus, León, Gto., México
| | - Elva Pérez-Luque
- Department of Medical Sciences, University of Guanajuato, León Campus, León, Gto., México
| | - Juan Manuel Malacara
- Department of Medical Sciences, University of Guanajuato, León Campus, León, Gto., México
| |
Collapse
|
19
|
SahBandar IN, Ndhlovu LC, Saiki K, Kohorn LB, Peterson MM, D'Antoni ML, Shiramizu B, Shikuma CM, Chow DC. Relationship between Circulating Inflammatory Monocytes and Cardiovascular Disease Measures of Carotid Intimal Thickness. J Atheroscler Thromb 2019; 27:441-448. [PMID: 31588100 PMCID: PMC7242227 DOI: 10.5551/jat.49791] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Aims: Cardiovascular disease (CVD) remains the leading cause of death worldwide despite improvements in the treatment of atherosclerosis, an inflammatory disease and major underlying cause of CVD. Monocytes, an innate immune cell type, are linked to CVD progression; however, given their heterogeneity, the association between distinct monocyte subsets and increased risk of CVD remains unclear. This study investigated the association between peripheral monocyte subpopulation numbers and carotid intima-media thickness (cIMT), a sensitive measure of CVD risk, in a cohort of adults recruited from the general population. Methods: We used clinical data and peripheral blood mononuclear cell (PBMC) specimens from 67 individuals. cIMT was measured by high-resolution, B-mode, ultrasound images of the right carotid artery. PBMCs were stained with conjugated monoclonal antibodies to define monocyte subpopulations based on CD14 and CD16 co-expressions into classical (CD14++CD16−), intermediate/inflammatory (CD14++CD16+), and non-classical/patrolling (CD14low/+CD16++) monocytes. Results: We found a higher intermediate monocyte count was significantly correlated with increased right common carotid artery (RCCA) and right carotid bifurcation (RBIF) intima-media thickness (IMT) (p = 0.004 and 0.006, respectively), even after adjusting for CVD-associated clinical data (p = 0.006 and 0.004, respectively). Conclusion: Our study demonstrated a strong correlation between inflammatory monocyte counts and cIMT. These results suggest that, in the general population, there is a relationship between intermediate monocyte expansion and elevated predictors for CVD risk, and intermediate monocytes may be involved in the development of atherosclerosis and metabolic diseases. Strategies targeting inflammatory monocytes may be needed to slow CVD progression.
Collapse
Affiliation(s)
- Ivo N SahBandar
- Hawai'i Center for AIDS, Department of Medicine, University of Hawaii John A. Burns School of Medicine.,Department of Tropical Medicine, University of Hawaii John A. Burns School of Medicine
| | - Lishomwa C Ndhlovu
- Hawai'i Center for AIDS, Department of Medicine, University of Hawaii John A. Burns School of Medicine.,Department of Tropical Medicine, University of Hawaii John A. Burns School of Medicine
| | - Katelyn Saiki
- Hawai'i Center for AIDS, Department of Medicine, University of Hawaii John A. Burns School of Medicine
| | - Lindsay B Kohorn
- Hawai'i Center for AIDS, Department of Medicine, University of Hawaii John A. Burns School of Medicine
| | - Mary Margaret Peterson
- Hawai'i Center for AIDS, Department of Medicine, University of Hawaii John A. Burns School of Medicine
| | - Michelle L D'Antoni
- Hawai'i Center for AIDS, Department of Medicine, University of Hawaii John A. Burns School of Medicine
| | - Bruce Shiramizu
- Hawai'i Center for AIDS, Department of Medicine, University of Hawaii John A. Burns School of Medicine.,Department of Tropical Medicine, University of Hawaii John A. Burns School of Medicine
| | - Cecilia M Shikuma
- Hawai'i Center for AIDS, Department of Medicine, University of Hawaii John A. Burns School of Medicine
| | - Dominic C Chow
- Hawai'i Center for AIDS, Department of Medicine, University of Hawaii John A. Burns School of Medicine
| |
Collapse
|
20
|
Monocyte Subsets, Stanford-A Acute Aortic Dissection, and Carotid Artery Stenosis: New Evidences. J Immunol Res 2019; 2019:9782594. [PMID: 31467936 PMCID: PMC6701364 DOI: 10.1155/2019/9782594] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 05/21/2019] [Accepted: 07/10/2019] [Indexed: 12/24/2022] Open
Abstract
Monocytes are a heterogeneous cell population distinguished into three subsets with distinctive phenotypic and functional properties: “classical” (CD14++CD16-), “intermediate” (CD14++CD16+), and “nonclassical” (CD14+CD16++). Monocyte subsets play a pivotal role in many inflammatory systemic diseases including atherosclerosis (ATS). Only a low number of studies evaluated monocyte behavior in patients affected by cardiovascular diseases, and data about their role in acute aortic dissection (AAD) are lacking. Thus, the aim of this study was to investigate CD14++CD16-, CD14++CD16+, and CD14+CD16++ cells in patients with Stanford-A AAD and in patients with carotid artery stenosis (CAS). Methods. 20 patients with carotid artery stenosis (CAS group), 17 patients with Stanford-A AAD (AAD group), and 17 subjects with traditional cardiovascular risk factors (RF group) were enrolled. Monocyte subset frequency was determined by flow cytometry. Results. Classical monocytes were significantly increased in the AAD group versus CAS and RF groups, whereas intermediate monocytes were significantly decreased in the AAD group versus CAS and RF groups. Conclusions. Results of this study identify in AAD patients a peculiar monocyte array that can partly explain depletion of T CD4+ lymphocyte subpopulations observed in patients affected by AAD.
Collapse
|
21
|
Rawther T, Tabet F. Biology, pathophysiology and current therapies that affect lipoprotein (a) levels. J Mol Cell Cardiol 2019; 131:1-11. [DOI: 10.1016/j.yjmcc.2019.04.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 03/22/2019] [Accepted: 04/09/2019] [Indexed: 12/11/2022]
|
22
|
Tabaei S, Tabaee SS. DNA methylation abnormalities in atherosclerosis. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2031-2041. [DOI: 10.1080/21691401.2019.1617724] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Samira Tabaei
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | |
Collapse
|
23
|
Ezhov M, Safarova M, Afanasieva O, Mitroshkin M, Matchin Y, Pokrovsky S. Matrix Metalloproteinase 9 as a Predictor of Coronary Atherosclerotic Plaque Instability in Stable Coronary Heart Disease Patients with Elevated Lipoprotein(a) Levels. Biomolecules 2019; 9:biom9040129. [PMID: 30934954 PMCID: PMC6523150 DOI: 10.3390/biom9040129] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 03/24/2019] [Accepted: 03/26/2019] [Indexed: 11/22/2022] Open
Abstract
We sought to investigate whether levels of matrix metalloproteinases (MMPs) and their inhibitors predict coronary atherosclerotic plaque instability, as assessed by intravascular ultrasound (IVUS) virtual histology during coronary angiography. Blood samples were collected before angiography in 32 subjects (mean age 56 ± 8 years) with stable coronary heart disease (CHD) and elevated lipoprotein(a) (Lp(a), 94 ± 35 mg/dL). Levels of high-sensitivity C-reactive protein (hsCRP), apolipoprotein B100 (apoB100), MMP-7, MMP-9, tissue inhibitor of metalloproteinases (TIMP)-1, and TIMP-2 were determined using commercially available enzyme-linked immunosorbent assay kits. Results. The morphology of a total of sixty coronary lesions was assessed by virtual histology IVUS imaging. Eleven (18%) plaques in nine (28%) patients were classified as plaques with an unstable phenotype or a thin-cap fibroatheroma. Age, low-density lipoprotein cholesterol, apoB100, MMP-7, and MMP-9 levels were positively associated with necrotic core volume. Conversely, there was a negative relationship between MMP-7 and -9 levels and fibrous and fibro-fatty tissue volume. Multivariate regression analysis revealed that MMP-9 is a strong independent predictor of atherosclerotic plaque instability in stable CHD patients. In stable CHD patients with elevated Lp(a), MMP-9 levels are positively associated with the size of the necrotic core of coronary atherosclerotic plaques.
Collapse
Affiliation(s)
- Marat Ezhov
- Federal State Budget Institution "National Cardiology Research Center" of Ministry of Health of the Russian Federation; 15A, 3d Cherepkovskaya street, 121552 Moscow, Russia.
| | - Maya Safarova
- Federal State Budget Institution "National Cardiology Research Center" of Ministry of Health of the Russian Federation; 15A, 3d Cherepkovskaya street, 121552 Moscow, Russia.
| | - Olga Afanasieva
- Federal State Budget Institution "National Cardiology Research Center" of Ministry of Health of the Russian Federation; 15A, 3d Cherepkovskaya street, 121552 Moscow, Russia.
| | - Maksim Mitroshkin
- Federal State Budget Institution "National Cardiology Research Center" of Ministry of Health of the Russian Federation; 15A, 3d Cherepkovskaya street, 121552 Moscow, Russia.
| | - Yuri Matchin
- Federal State Budget Institution "National Cardiology Research Center" of Ministry of Health of the Russian Federation; 15A, 3d Cherepkovskaya street, 121552 Moscow, Russia.
| | - Sergei Pokrovsky
- Federal State Budget Institution "National Cardiology Research Center" of Ministry of Health of the Russian Federation; 15A, 3d Cherepkovskaya street, 121552 Moscow, Russia.
| |
Collapse
|
24
|
Del Porto F, Cifani N, Proietta M, Dezi T, Panzera C, Ficarelli R, Taurino M. Inflammation and immune response in carotid artery stenosis. ITALIAN JOURNAL OF VASCULAR AND ENDOVASCULAR SURGERY 2019. [DOI: 10.23736/s1824-4777.18.01385-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
25
|
Leenders GJ, Smeets MB, van den Boomen M, Berben M, Nabben M, van Strijp D, Strijkers GJ, Prompers JJ, Arslan F, Nicolay K, Vandoorne K. Statins Promote Cardiac Infarct Healing by Modulating Endothelial Barrier Function Revealed by Contrast-Enhanced Magnetic Resonance Imaging. Arterioscler Thromb Vasc Biol 2017; 38:186-194. [PMID: 29146749 DOI: 10.1161/atvbaha.117.310339] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 10/24/2017] [Indexed: 01/13/2023]
Abstract
OBJECTIVE The endothelium has a crucial role in wound healing, acting as a barrier to control transit of leukocytes. Endothelial barrier function is impaired in atherosclerosis preceding myocardial infarction (MI). Besides lowering lipids, statins modulate endothelial function. Here, we noninvasively tested whether statins affect permeability at the inflammatory (day 3) and the reparative (day 7) phase of infarct healing post-MI using contrast-enhanced cardiac magnetic resonance imaging (MRI). APPROACH AND RESULTS Noninvasive permeability mapping by MRI after MI in C57BL/6, atherosclerotic ApoE-/-, and statin-treated ApoE-/- mice was correlated to subsequent left ventricular outcome by structural and functional cardiac MRI. Ex vivo histology, flow cytometry, and quantitative polymerase chain reaction were performed on infarct regions. Increased vascular permeability at ApoE-/- infarcts was observed compared with C57BL/6 infarcts, predicting enhanced left ventricular dilation at day 21 post-MI by MRI volumetry. Statin treatment improved vascular barrier function at ApoE-/- infarcts, indicated by reduced permeability. The infarcted tissue of ApoE-/- mice 3 days post-MI displayed an unbalanced Vegfa(vascular endothelial growth factor A)/Angpt1 (angiopoetin-1) expression ratio (explaining leakage-prone vessels), associated with higher amounts of CD45+ leukocytes and inflammatory LY6Chi monocytes. Statins reversed the unbalanced Vegfa/Angpt1 expression, normalizing endothelial barrier function at the infarct and blocking the augmented recruitment of inflammatory leukocytes in statin-treated ApoE-/- mice. CONCLUSIONS Statins lowered permeability and reduced the transit of unfavorable inflammatory leukocytes into the infarcted tissue, consequently improving left ventricular outcome.
Collapse
Affiliation(s)
- Geert J Leenders
- From the Department of Biomedical Engineering, Biomedical NMR, Eindhoven University of Technology, The Netherlands (G.J.L., M.v.d.B., M.N., G.J.S., J.J.P., K.N., K.V.); Laboratory of Experimental Cardiology (M.B.S.) and Department of Cardiology (F.A.), University Medical Center Utrecht, The Netherlands; Department Precision and Decentralized Diagnostics, Philips Research Eindhoven, The Netherlands (M.B., D.v.S.); Biomedical Engineering and Physics, Academic Medical Center, Amsterdam, The Netherlands (G.J.S.); and Department of Cardiology, St. Antonius Hospital Nieuwegein, The Netherlands (F.A.)
| | - Mirjam B Smeets
- From the Department of Biomedical Engineering, Biomedical NMR, Eindhoven University of Technology, The Netherlands (G.J.L., M.v.d.B., M.N., G.J.S., J.J.P., K.N., K.V.); Laboratory of Experimental Cardiology (M.B.S.) and Department of Cardiology (F.A.), University Medical Center Utrecht, The Netherlands; Department Precision and Decentralized Diagnostics, Philips Research Eindhoven, The Netherlands (M.B., D.v.S.); Biomedical Engineering and Physics, Academic Medical Center, Amsterdam, The Netherlands (G.J.S.); and Department of Cardiology, St. Antonius Hospital Nieuwegein, The Netherlands (F.A.)
| | - Maaike van den Boomen
- From the Department of Biomedical Engineering, Biomedical NMR, Eindhoven University of Technology, The Netherlands (G.J.L., M.v.d.B., M.N., G.J.S., J.J.P., K.N., K.V.); Laboratory of Experimental Cardiology (M.B.S.) and Department of Cardiology (F.A.), University Medical Center Utrecht, The Netherlands; Department Precision and Decentralized Diagnostics, Philips Research Eindhoven, The Netherlands (M.B., D.v.S.); Biomedical Engineering and Physics, Academic Medical Center, Amsterdam, The Netherlands (G.J.S.); and Department of Cardiology, St. Antonius Hospital Nieuwegein, The Netherlands (F.A.)
| | - Monique Berben
- From the Department of Biomedical Engineering, Biomedical NMR, Eindhoven University of Technology, The Netherlands (G.J.L., M.v.d.B., M.N., G.J.S., J.J.P., K.N., K.V.); Laboratory of Experimental Cardiology (M.B.S.) and Department of Cardiology (F.A.), University Medical Center Utrecht, The Netherlands; Department Precision and Decentralized Diagnostics, Philips Research Eindhoven, The Netherlands (M.B., D.v.S.); Biomedical Engineering and Physics, Academic Medical Center, Amsterdam, The Netherlands (G.J.S.); and Department of Cardiology, St. Antonius Hospital Nieuwegein, The Netherlands (F.A.)
| | - Miranda Nabben
- From the Department of Biomedical Engineering, Biomedical NMR, Eindhoven University of Technology, The Netherlands (G.J.L., M.v.d.B., M.N., G.J.S., J.J.P., K.N., K.V.); Laboratory of Experimental Cardiology (M.B.S.) and Department of Cardiology (F.A.), University Medical Center Utrecht, The Netherlands; Department Precision and Decentralized Diagnostics, Philips Research Eindhoven, The Netherlands (M.B., D.v.S.); Biomedical Engineering and Physics, Academic Medical Center, Amsterdam, The Netherlands (G.J.S.); and Department of Cardiology, St. Antonius Hospital Nieuwegein, The Netherlands (F.A.)
| | - Dianne van Strijp
- From the Department of Biomedical Engineering, Biomedical NMR, Eindhoven University of Technology, The Netherlands (G.J.L., M.v.d.B., M.N., G.J.S., J.J.P., K.N., K.V.); Laboratory of Experimental Cardiology (M.B.S.) and Department of Cardiology (F.A.), University Medical Center Utrecht, The Netherlands; Department Precision and Decentralized Diagnostics, Philips Research Eindhoven, The Netherlands (M.B., D.v.S.); Biomedical Engineering and Physics, Academic Medical Center, Amsterdam, The Netherlands (G.J.S.); and Department of Cardiology, St. Antonius Hospital Nieuwegein, The Netherlands (F.A.)
| | - Gustav J Strijkers
- From the Department of Biomedical Engineering, Biomedical NMR, Eindhoven University of Technology, The Netherlands (G.J.L., M.v.d.B., M.N., G.J.S., J.J.P., K.N., K.V.); Laboratory of Experimental Cardiology (M.B.S.) and Department of Cardiology (F.A.), University Medical Center Utrecht, The Netherlands; Department Precision and Decentralized Diagnostics, Philips Research Eindhoven, The Netherlands (M.B., D.v.S.); Biomedical Engineering and Physics, Academic Medical Center, Amsterdam, The Netherlands (G.J.S.); and Department of Cardiology, St. Antonius Hospital Nieuwegein, The Netherlands (F.A.)
| | - Jeanine J Prompers
- From the Department of Biomedical Engineering, Biomedical NMR, Eindhoven University of Technology, The Netherlands (G.J.L., M.v.d.B., M.N., G.J.S., J.J.P., K.N., K.V.); Laboratory of Experimental Cardiology (M.B.S.) and Department of Cardiology (F.A.), University Medical Center Utrecht, The Netherlands; Department Precision and Decentralized Diagnostics, Philips Research Eindhoven, The Netherlands (M.B., D.v.S.); Biomedical Engineering and Physics, Academic Medical Center, Amsterdam, The Netherlands (G.J.S.); and Department of Cardiology, St. Antonius Hospital Nieuwegein, The Netherlands (F.A.)
| | - Fatih Arslan
- From the Department of Biomedical Engineering, Biomedical NMR, Eindhoven University of Technology, The Netherlands (G.J.L., M.v.d.B., M.N., G.J.S., J.J.P., K.N., K.V.); Laboratory of Experimental Cardiology (M.B.S.) and Department of Cardiology (F.A.), University Medical Center Utrecht, The Netherlands; Department Precision and Decentralized Diagnostics, Philips Research Eindhoven, The Netherlands (M.B., D.v.S.); Biomedical Engineering and Physics, Academic Medical Center, Amsterdam, The Netherlands (G.J.S.); and Department of Cardiology, St. Antonius Hospital Nieuwegein, The Netherlands (F.A.)
| | - Klaas Nicolay
- From the Department of Biomedical Engineering, Biomedical NMR, Eindhoven University of Technology, The Netherlands (G.J.L., M.v.d.B., M.N., G.J.S., J.J.P., K.N., K.V.); Laboratory of Experimental Cardiology (M.B.S.) and Department of Cardiology (F.A.), University Medical Center Utrecht, The Netherlands; Department Precision and Decentralized Diagnostics, Philips Research Eindhoven, The Netherlands (M.B., D.v.S.); Biomedical Engineering and Physics, Academic Medical Center, Amsterdam, The Netherlands (G.J.S.); and Department of Cardiology, St. Antonius Hospital Nieuwegein, The Netherlands (F.A.)
| | - Katrien Vandoorne
- From the Department of Biomedical Engineering, Biomedical NMR, Eindhoven University of Technology, The Netherlands (G.J.L., M.v.d.B., M.N., G.J.S., J.J.P., K.N., K.V.); Laboratory of Experimental Cardiology (M.B.S.) and Department of Cardiology (F.A.), University Medical Center Utrecht, The Netherlands; Department Precision and Decentralized Diagnostics, Philips Research Eindhoven, The Netherlands (M.B., D.v.S.); Biomedical Engineering and Physics, Academic Medical Center, Amsterdam, The Netherlands (G.J.S.); and Department of Cardiology, St. Antonius Hospital Nieuwegein, The Netherlands (F.A.).
| |
Collapse
|
26
|
The effects of vitamin E or lipoic acid supplementation on oxyphytosterols in subjects with elevated oxidative stress: a randomized trial. Sci Rep 2017; 7:15288. [PMID: 29127425 PMCID: PMC5681676 DOI: 10.1038/s41598-017-15615-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 10/27/2017] [Indexed: 12/19/2022] Open
Abstract
Despite increased serum plant sterol concentrations after consumption of plant sterol enriched margarines, plasma oxyphytosterol concentrations were not increased in healthy subjects. Here, we assessed plasma oxyphytosterol concentrations and whether they are affected by antioxidants in subjects with elevated oxidative stress. Twenty subjects with impaired glucose tolerance (IGT) or type 2 diabetes (DM2) consumed for 4 weeks placebo, vitamin E (804 mg/d) or lipoic acid capsules (600 mg/d). Plasma and blood cell oxyphytosterol and oxycholesterol concentrations were determined in butylated hydroxytoluene-enriched EDTA plasma via GC-MS. Also, markers reflecting oxidative stress and antioxidant capacity were measured. Plasma oxycampesterol and oxysitosterol concentrations were 122% and 83% higher in IGT or DM2 subjects than in healthy subjects, as determined in an earlier study. Vitamin E or lipoic acid supplementation did not reduce plasma oxyphytosterol and oxycholesterol concentrations, or other markers reflecting oxidative stress or antioxidative capacity. Concentrations of different oxyphytosterols correlated within plasma, and within red blood cells and platelets. However, plasma and blood cell oxyphytosterol levels did not correlate. Although plasma oxyphytosterol concentrations are higher in IGT or DM2 subjects than in healthy subjects, 4-weeks vitamin E or lipoic acid supplementation does not lower plasma oxycholesterol or oxyphytosterol concentrations.
Collapse
|
27
|
Jellinghaus S, Reich C, Schatz U, Tselmin S, Ibrahim K, Pfluecke C, Schauer A, Bornstein SR, Hohenstein B, Strasser RH, Julius U, Poitz DM. Lipoprotein apheresis influences monocyte subpopulations. ATHEROSCLEROSIS SUPP 2017; 30:108-114. [PMID: 29096825 DOI: 10.1016/j.atherosclerosissup.2017.05.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND Monocytes can be differentiated into subpopulations depending on their expression profile of CD14 and CD16. CD16-positive monocytes are associated with coronary artery disease. Up to now, no data exist about the effect of lipoprotein apheresis (LA) on the distribution of monocyte subpopulations. METHODS 80 patients who underwent LA at the University Hospital Dresden were included in the study. 8 out of the 80 LA patients received LA for the first time at the time point of blood analysis. Six different methods of LA were used (H.E.L.P. n = 8; Liposorber D n = 10; LF n = 14; DALI n = 17; MONET n = 11; Therasorb® LDL n = 12). Blood samples were taken immediately before and after LA and analyzed for CD14 and CD16 expression on monocytes. A total of 42 patients with cardiovascular risk factors but no indication for LA served as control group. RESULTS The composition of monocyte-population was analyzed in regard to the 3 subpopulations. After LA, an increase in classical monocytes (CD14++CD16-) (93.3% vs. 93.9%, p < 0.01) and a decrease in non-classical monocytes (CD14+CD16+) (1.5% vs 1.0%; p < 0.001) were observed. LA did not change the amount of intermediate monocytes (CD14++CD16+) (5.3% vs. 5.1%). Two methods (MONET and Therasorb® LDL) did not influence the distribution of monocyte subpopulations. Interestingly, patients with LDL-C above 2.5 mmol/l prior LA showed increased amounts of intermediate monocytes. CONCLUSION The distribution of monocyte populations is influenced by LA but depends on the distinct method of LA. Influences of LA were mainly observed in the content of classical and non-classical monocytes, whereas the intermediate monocyte population remained unaltered by LA.
Collapse
Affiliation(s)
- S Jellinghaus
- Internal Medicine and Cardiology, Heart Center Dresden, University Hospital at the Technische Universität, Dresden, Germany
| | - C Reich
- Internal Medicine and Cardiology, Heart Center Dresden, University Hospital at the Technische Universität, Dresden, Germany
| | - U Schatz
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - S Tselmin
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - K Ibrahim
- Internal Medicine and Cardiology, Heart Center Dresden, University Hospital at the Technische Universität, Dresden, Germany
| | - C Pfluecke
- Internal Medicine and Cardiology, Heart Center Dresden, University Hospital at the Technische Universität, Dresden, Germany
| | - A Schauer
- Internal Medicine and Cardiology, Heart Center Dresden, University Hospital at the Technische Universität, Dresden, Germany
| | - S R Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - B Hohenstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - R H Strasser
- Internal Medicine and Cardiology, Heart Center Dresden, University Hospital at the Technische Universität, Dresden, Germany
| | - U Julius
- Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - D M Poitz
- Internal Medicine and Cardiology, Heart Center Dresden, University Hospital at the Technische Universität, Dresden, Germany.
| |
Collapse
|
28
|
Altered leukocyte distribution under hypercholesterolemia: A cross-sectional study in children with familial hypercholesterolemia. Atherosclerosis 2016; 256:67-74. [PMID: 28024183 DOI: 10.1016/j.atherosclerosis.2016.11.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 11/29/2016] [Accepted: 11/30/2016] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND AIMS Children with familial hypercholesterolemia (FH) have elevated LDL cholesterol from the first year of life, and represent a model of early-stage atherosclerosis. Data suggest that adults with FH have alterations in circulating monocyte subpopulations towards a more pro-inflammatory phenotype, but it is not known whether FH children have similar perturbations. In addition, there are no data on the distribution of lymphocyte subpopulations in FH children. The objective of the present study was to characterize the distributions of circulating monocyte and lymphocyte subpopulations in children with FH and healthy, normocholesterolemic children. METHODS Using flow cytometry analysis, we analyzed whole blood B- and T-cell subpopulations and monocyte subpopulations in FH (n = 23) and healthy (n = 20) children. Moreover, we measured serum markers of leukocyte and endothelial cell activation using EIA. RESULTS We found that FH children had monocytosis as well as a shift in the monocyte subpopulations. This shift was characterized by higher circulating pro-inflammatory and non-classical monocytes, and lower levels of classical monocytes, and seemed to be present only in FH children with low HDL cholesterol (HDL-C, below 1.3 mmol/L). Additionally, monocytes expressing CD18 and serum E-selectin were higher in FH children, in particular FH children with low HDL-C. CONCLUSIONS FH children with low HDL-C had monocytosis as well as a shift in monocyte subpopulations towards a more pro-inflammatory phenotype. Our results suggest activation of monocytes at a very early stage of atherosclerosis in humans.
Collapse
|
29
|
Dong J, Liang YZ, Zhang J, Wu LJ, Wang S, Hua Q, Yan YX. Potential Role of Lipometabolism-Related MicroRNAs in Peripheral Blood Mononuclear Cells as Biomarkers for Coronary Artery Disease. J Atheroscler Thromb 2016; 24:430-441. [PMID: 27629254 PMCID: PMC5392481 DOI: 10.5551/jat.35923] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aim: To explore the relationship between lipometabolism-related microRNAs (miRNAs) in peripheral blood mononuclear cells (PBMCs) and the presence of coronary artery disease (CAD). Methods: In the present study, 161 stable CAD patients and 149 health controls were enrolled. The expression levels of seven miRNAs (miR-21, miR-24, miR-29a, miR-33a, miR-34a, miR-103a, and miR-122) in PBMCs were qualified by quantitative real-time polymerase chain reaction (qRT-PCR). The miRNA markers that showed significant difference between the two groups were used for further analysis. The risk of miRNA contributing to the presence of CAD was estimated by univariate and multivariate logistic regression models. The area under the receiver operating characteristic curve (AUC) was used to evaluate diagnostic accuracy. Results: The expression levels of miR-24, miR-33a, miR-103a, and miR-122 in PBMCs were significantly increased in CAD patients compared with controls and were significantly correlated with blood lipids in both CAD patients and controls. The increased levels of miR-24 (adjusted OR = 1.32, 95% CI 1.07–1.62, P = 0.009), miR-33a (adjusted OR = 1.57, 95% CI 1.35–1.81, P < 0.001), miR-103a (adjusted OR = 1.01, 95% CI 1.01–1.02, P < 0.001), and miR-122 (adjusted OR = 1.03, 95% CI 1.01–1.04, P < 0.001) were associated with risk of CAD. We identified a miRNA panel (miR-24, miR-33, miR-103a, and miR-122) that provided a high diagnostic accuracy of CAD (AUC= 0.911, 95% CI 0.880–0.942). Conclusion: The increased expression levels of miR-24, miR-33a, miR-103a, and miR-122 in PBMCs are associated with risk of CAD. A panel of the four miRNAs has considerable clinical value in diagnosing stable CAD.
Collapse
Affiliation(s)
- Jing Dong
- Health Medical Examination Center, Xuanwu Hospital, Capital Medical University
| | | | | | | | | | | | | |
Collapse
|
30
|
Wildgruber M, Czubba M, Aschenbrenner T, Wendorff H, Hapfelmeier A, Glinzer A, Schiemann M, Zimmermann A, Eckstein HH, Berger H, Wohlgemuth WA, Meier R, Libby P, Zernecke A. Increased intermediate CD14 ++CD16 ++ monocyte subset levels associate with restenosis after peripheral percutaneous transluminal angioplasty. Atherosclerosis 2016; 253:128-134. [PMID: 27615596 DOI: 10.1016/j.atherosclerosis.2016.09.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 08/31/2016] [Accepted: 09/01/2016] [Indexed: 11/17/2022]
Abstract
BACKGROUND AND AIMS We aimed at studying the association of three major human monocyte subsets after percutaneous transluminal angioplasty (PTA) in patients with femoropopliteal disease. METHODS We prospectively studied 67 sequential patients (40 male, 27 female; mean age 71 ± 11 years) treated with femoropopliteal angioplasty. Multi-color flow cytometry characterized monocyte subsets from venous blood for expression of CD14 and CD16 and intracellular myeloperoxidase (MPO) prior to, and 3, 6 and 12 months post PTA. Analyses tested associations between monocyte subsets and risk for restenosis. RESULTS 16/67 patients (24%) developed restenosis within 12 months after PTA. Patients with hyperlipidemia had increased risk for restenosis (HR = 1.7, 95% CI 0.7-2.9, p = 0.001). Increased baseline monocytes associated with an increased risk of late restenosis (HR = 4.9, 95% CI: 1.3-18.6, p = 0.047). CD14++CD16++ 'intermediate' monocytes assessed at baseline, and after 3, 6, and 12 months significantly associated with the risk for subsequent restenosis: HR = 3.9 (95% CI: 2.4-6.5, p = 0.029), HR = 5.7 (95% CI = 0.7-44.7, p = 0.013), HR = 6.5 (95% CI: 2.5-16.9, p = 0.001) and HR = 1.5 (95% CI = 1.4-15.5 p = 0.001), respectively. Moreover, the probability for freedom of restenosis decreased with increased levels of intermediate subsets at 12 months after PTA. Additionally, intracellular MPO expression in CD14++CD16++ measured at 3, 6 and 12 months associated with an increased restenosis risk (HR = 1.5, 95% CI: 0.8-2.1, p = 0.214, HR = 1.9, 95% CI: 1.0-2.3 p = 0.051 and HR = 1.4, 95% CI: 1.0-1.8, p = 0.052). CONCLUSIONS Our results imply altered innate immunity after angioplasty. Elevated CD14++CD16++ intermediate monocyte frequencies and increased MPO expression may identify individuals at heightened risk for restenosis.
Collapse
Affiliation(s)
- Moritz Wildgruber
- Institut für diagnostische und interventionelle Radiologie, Klinikum rechts der Isar, Technische Universität München, Germany; Institut für klinische Radiologie, Universitätsklinikum Münster, Germany.
| | - Maria Czubba
- Institut für diagnostische und interventionelle Radiologie, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Teresa Aschenbrenner
- Institut für diagnostische und interventionelle Radiologie, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Heiko Wendorff
- Klinik für vaskuläre und endovaskuläre Chirurgie, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Alexander Hapfelmeier
- Institut für Medizinische Statistik und Epidemiologie, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Almut Glinzer
- Institut für diagnostische und interventionelle Radiologie, Klinikum rechts der Isar, Technische Universität München, Germany; Klinik für vaskuläre und endovaskuläre Chirurgie, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Matthias Schiemann
- Institut für medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, Germany; Klinische Kooperationsgemeinschaft, "Immunmonitoring", Helmholtz Zentrum München und Technische Universität München, München, Germany
| | - Alexander Zimmermann
- Klinik für vaskuläre und endovaskuläre Chirurgie, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Hans-Henning Eckstein
- Klinik für vaskuläre und endovaskuläre Chirurgie, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Hermann Berger
- Institut für diagnostische und interventionelle Radiologie, Klinikum rechts der Isar, Technische Universität München, Germany
| | | | - Reinhard Meier
- Klinik für diagnostische und interventionelle Radiologie, Universitätsklinikum Ulm, Germany
| | - Peter Libby
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Alma Zernecke
- Institut für Klinische Biochemie und Pathobiochemie, Universitätsklinikum Würzburg, Germany
| |
Collapse
|
31
|
Thaler B, Hohensinner PJ, Krychtiuk KA, Matzneller P, Koller L, Brekalo M, Maurer G, Huber K, Zeitlinger M, Jilma B, Wojta J, Speidl WS. Differential in vivo activation of monocyte subsets during low-grade inflammation through experimental endotoxemia in humans. Sci Rep 2016; 6:30162. [PMID: 27444882 PMCID: PMC4957086 DOI: 10.1038/srep30162] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 06/28/2016] [Indexed: 02/07/2023] Open
Abstract
Human monocytes are a heterogeneous cell population, which can be divided into a classical (CD14++CD16−), a non-classical (CD14+CD16+), and an intermediate (CD14++CD16+) subset. We hypothesized that low-grade inflammation may differentially affect monocyte subsets. We used a human lipopolysaccharide (LPS) infusion model to mimic low-grade inflammation to identify, which monocyte subsets are preferentially activated under these conditions. Monocyte subsets were identified by staining for CD14 and CD16, activation status of monocytes was analyzed by staining for CD11b and a novel in situ mRNA hybridization approach to detect IL-6 and IL-8 specific mRNA at the single-cell level by flow cytometry. After LPS challenge, cell numbers of monocyte subsets dropped after 2 h with cell numbers recovering after 6 h. Distribution of monocyte subsets was skewed dramatically towards the intermediate subset after 24 h. Furthermore, intermediate monocytes displayed the largest increase of CD11b expression after 2 h. Finally, IL-6 and IL-8 mRNA levels increased in intermediate and non-classical monocytes after 6 h whereas these mRNA levels in classical monocytes changed only marginally. In conclusion, our data indicates that the main responding subset of monocytes to standardized low-grade inflammation induced by LPS in humans is the CD14++CD16+ intermediate subset followed by the CD14+CD16+ non-classical monocyte subset. Circulating classical monocytes showed comparably less reaction to LPS challenge in vivo.
Collapse
Affiliation(s)
- B Thaler
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - P J Hohensinner
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - K A Krychtiuk
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria
| | - P Matzneller
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - L Koller
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - M Brekalo
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - G Maurer
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - K Huber
- 3rd Medical Department for Cardiology and Emergency Medicine, Wilhelminen Hospital, Vienna, Austria
| | - M Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - B Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - J Wojta
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria.,Core Facilities, Medical University of Vienna, Vienna, Austria
| | - W S Speidl
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
32
|
Weber C, Shantsila E, Hristov M, Caligiuri G, Guzik T, Heine GH, Hoefer IE, Monaco C, Peter K, Rainger E, Siegbahn A, Steffens S, Wojta J, Lip GYH. Role and analysis of monocyte subsets in cardiovascular disease. Joint consensus document of the European Society of Cardiology (ESC) Working Groups "Atherosclerosis & Vascular Biology" and "Thrombosis". Thromb Haemost 2016; 116:626-37. [PMID: 27412877 DOI: 10.1160/th16-02-0091] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 06/02/2016] [Indexed: 12/21/2022]
Abstract
Monocytes as cells of the innate immunity are prominently involved in the development of atherosclerotic lesions. The heterogeneity of blood monocytes has widely been acknowledged by accumulating experimental and clinical data suggesting a differential, subset-specific contribution of the corresponding subpopulations to the pathology of cardiovascular and other diseases. This document re-evaluates current nomenclature and summarises key findings on monocyte subset biology to propose a consensus statement about phenotype, separation and quantification of the individual subsets.
Collapse
Affiliation(s)
- Christian Weber
- Dr. Christian Weber, LMU Munich - Cardiovascular Prevention, Pettenkoferstr. 9, 80336 Munich, Germany, Tel.: +49 89 4400 54350, Fax: +49 89 4400 54352, E-mail:
| | | | - Michael Hristov
- Dr. Michael Hristov, LMU Munich - Cardiovascular Prevention, Pettenkoferstr. 9, 80336 Munich, Germany, Tel.: +49 89 4400 54350, Fax: +49 89 4400 54352, E-mail:
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Hai Z, Zuo W. Aberrant DNA methylation in the pathogenesis of atherosclerosis. Clin Chim Acta 2016; 456:69-74. [DOI: 10.1016/j.cca.2016.02.026] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/24/2016] [Accepted: 02/28/2016] [Indexed: 11/26/2022]
|
34
|
Gijsberts CM, Ellenbroek GH, Ten Berg MJ, Huisman A, van Solinge WW, Asselbergs FW, den Ruijter HM, Pasterkamp G, de Kleijn DP, Hoefer IE. Routinely analyzed leukocyte characteristics improve prediction of mortality after coronary angiography. Eur J Prev Cardiol 2015; 23:1211-20. [PMID: 26643521 DOI: 10.1177/2047487315621832] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 11/20/2015] [Indexed: 12/25/2022]
Abstract
BACKGROUND Inflammation and leukocyte infiltration are hallmarks of atherosclerosis. Clinically routine hematology analyzers mostly perform an entire differential blood count by default, irrespective of the requested parameter. We hypothesize that these normally unreported leukocyte characteristics associate with coronary artery disease (CAD) severity and can improve prediction of mortality in coronary angiography patients. METHODS We studied coronary angiography patients suspected of CAD (n = 1015) from the Utrecht Coronary Biobank cohort. Leukocyte characteristics were routinely assessed in blood drawn directly prior to angiography using an automated hematology analyzer and extracted from the Utrecht patient oriented database (UPOD) database. Patients were followed up for a median duration of 805 days, during which 65 patients died. We evaluated the association of leukocyte characteristics with synergy between PCI with taxus and cardiac surgery (SYNTAX) score as a measure of CAD severity, all-cause and cardiovascular mortality and major adverse cardiovascular events (MACEs). In order to determine the improvement of risk prediction, we calculated continuous net reclassification improvement (cNRI) and integrated discrimination improvement (IDI). RESULTS Monocyte percentage showed strong independent predictive value for all-cause mortality (hazard ratio (HR) 1.44 (1.19-1.74), p < 0.001), and the monocyte-to-lymphocyte ratio performed best for cardiovascular mortality (HR 1.42 (1.11-1.81), p = 0.005). The cNRIs and IDIs of leukocyte characteristics for all-cause mortality confirmed the improvement in mortality risk prediction. No significantly predictive leukocyte characteristics were found for MACEs. CONCLUSION Readily available yet unreported leukocyte characteristics from routine hematology analyzers significantly improved prediction of mortality in coronary angiography patients on top of clinical characteristics.
Collapse
Affiliation(s)
- Crystel M Gijsberts
- Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, The Netherlands ICIN-Netherlands Heart Institute, the Netherlands
| | | | - Maarten J Ten Berg
- Department of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Albert Huisman
- Department of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wouter W van Solinge
- Department of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Folkert W Asselbergs
- Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, Utrecht, The Netherlands Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London, United Kingdom
| | - Hester M den Ruijter
- Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gerard Pasterkamp
- Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, The Netherlands Department of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Dominique Pv de Kleijn
- Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, The Netherlands ICIN-Netherlands Heart Institute, the Netherlands Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore Cardiovascular Research Institute (CVRI), National University Heart Centre (NUHCS), National University Health System, Singapore
| | - Imo E Hoefer
- Experimental Cardiology Laboratory, University Medical Center Utrecht, Utrecht, The Netherlands Department of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|