101
|
Babur Ö, Melrose AR, Cunliffe JM, Klimek J, Pang J, Sepp ALI, Zilberman-Rudenko J, Tassi Yunga S, Zheng T, Parra-Izquierdo I, Minnier J, McCarty OJT, Demir E, Reddy AP, Wilmarth PA, David LL, Aslan JE. Phosphoproteomic quantitation and causal analysis reveal pathways in GPVI/ITAM-mediated platelet activation programs. Blood 2020; 136:2346-2358. [PMID: 32640021 PMCID: PMC7702475 DOI: 10.1182/blood.2020005496] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/05/2020] [Indexed: 02/07/2023] Open
Abstract
Platelets engage cues of pending vascular injury through coordinated adhesion, secretion, and aggregation responses. These rapid, progressive changes in platelet form and function are orchestrated downstream of specific receptors on the platelet surface and through intracellular signaling mechanisms that remain systematically undefined. This study brings together cell physiological and phosphoproteomics methods to profile signaling mechanisms downstream of the immunotyrosine activation motif (ITAM) platelet collagen receptor GPVI. Peptide tandem mass tag (TMT) labeling, sample multiplexing, synchronous precursor selection (SPS), and triple stage tandem mass spectrometry (MS3) detected >3000 significant (false discovery rate < 0.05) phosphorylation events on >1300 proteins over conditions initiating and progressing GPVI-mediated platelet activation. With literature-guided causal inference tools, >300 site-specific signaling relations were mapped from phosphoproteomics data among key and emerging GPVI effectors (ie, FcRγ, Syk, PLCγ2, PKCδ, DAPP1). Through signaling validation studies and functional screening, other less-characterized targets were also considered within the context of GPVI/ITAM pathways, including Ras/MAPK axis proteins (ie, KSR1, SOS1, STAT1, Hsp27). Highly regulated GPVI/ITAM targets out of context of curated knowledge were also illuminated, including a system of >40 Rab GTPases and associated regulatory proteins, where GPVI-mediated Rab7 S72 phosphorylation and endolysosomal maturation were blocked by TAK1 inhibition. In addition to serving as a model for generating and testing hypotheses from omics datasets, this study puts forth a means to identify hemostatic effectors, biomarkers, and therapeutic targets relevant to thrombosis, vascular inflammation, and other platelet-associated disease states.
Collapse
Affiliation(s)
- Özgün Babur
- Department of Molecular and Medical Genetics
- Computational Biology Program
| | | | | | | | | | | | | | | | | | | | | | | | - Emek Demir
- Department of Molecular and Medical Genetics
- Computational Biology Program
| | | | | | - Larry L David
- Proteomics Shared Resource
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR
| | - Joseph E Aslan
- Knight Cardiovascular Institute
- Department of Biomedical Engineering
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR
| |
Collapse
|
102
|
Konkoth A, Saraswat R, Dubrou C, Sabatier F, Leroyer AS, Lacroix R, Duchez AC, Dignat-George F. Multifaceted role of extracellular vesicles in atherosclerosis. Atherosclerosis 2020; 319:121-131. [PMID: 33261815 DOI: 10.1016/j.atherosclerosis.2020.11.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/13/2020] [Accepted: 11/05/2020] [Indexed: 12/19/2022]
Abstract
Extracellular vesicles (EVs) are small vesicles released by the majority of cells in response to cell activation or death stimuli. They are grouped as small EVs or exosomes, large EVs such as microvesicles (MVs) and apoptotic bodies, resulting from distinct mechanisms of generation. EVs are released into the extracellular space, in most human biological fluids and tissues, including atherosclerotic plaques. They transport complex cargo of bioactive molecules, including proteins, lipids and genetic material and are therefore involved in pathophysiological pathways of cell-cell communication. Indeed, EVs are involved in several processes such as inflammation, coagulation, vascular dysfunction, angiogenesis and senescence, contributing to the initiation and progression of atherothrombotic diseases. Consequently, they behave as a determinant of atherosclerotic plaque vulnerability leading to major cardiovascular disorders. Over the last decade, the field of EVs research has grown, highlighting their involvement in atherosclerosis. However, limitations in both detection methodologies and standardisation have hindered implementation of EVs in the clinical settings. This review summarizes the effect of EVs in atherosclerosis development, progression and severity, with specific attention devoted to their ambivalent roles in senescence and hemostasis. This review will also highlight the role of MVs as multifaceted messengers, able to promote or to attenuate atherosclerosis progression. Finally, we will discuss the main technical challenges and prerequisites of standardization for driving EVs to the clinics and delineate their relevance as emergent biomarkers and innovative therapeutic approaches in atherosclerosis.
Collapse
Affiliation(s)
- Akhil Konkoth
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France
| | - Ronald Saraswat
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France
| | - Cléa Dubrou
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France; Department of Hematology and Vascular Biology, CHU La Conception, APHM, Marseille, France
| | - Florence Sabatier
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France; Department of Hematology and Vascular Biology, CHU La Conception, APHM, Marseille, France
| | | | - Romaric Lacroix
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France; Department of Hematology and Vascular Biology, CHU La Conception, APHM, Marseille, France
| | | | - Francoise Dignat-George
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France; Department of Hematology and Vascular Biology, CHU La Conception, APHM, Marseille, France
| |
Collapse
|
103
|
Bourassa KA, Postolache TT, Dagdag A, Fuchs D, Okusaga OO. Plasma soluble P-selectin correlates with triglycerides and nitrite in overweight/obese patients with schizophrenia. Pteridines 2020; 31:61-67. [PMID: 32982068 PMCID: PMC7518413 DOI: 10.1515/pteridines-2020-0012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background: Soluble P-selectin (sP-selectin) is associated with risk factors for cardiovascular disease (CVD) but this association has not been evaluated in patients with schizophrenia. This study primarily evaluated the association of sP-selectin with plasma lipids and nitrite (NO2−) respectively in overweight/obese adults with schizophrenia. Methods: One-hundred and six patients with schizophrenia (mean age 32.9 years; 71.60% male) were recruited from a psychiatric hospital. Participants completed a structured interview and provided a fasting blood sample. Body mass index (BMI) was used to divide the sample into normal weight and overweight/obese groups. Pearson’s and partial correlation coefficients (controlling for age, sex, race, education, and inflammation) were calculated to examine the association of sP-selectin with plasma lipids, and NO2− in the overweight/obese patients (primary analysis), as well as in the normal weight patients and the total sample (exploratory analyses). Results: After controlling for potential confounders, sP-selectin positively correlated with triglycerides (r = 0.38, p = 0.01) and NO2− (r = 0.40, p < 0.01) in the overweight/obese group only. Conclusions: Future longitudinal studies should evaluate the utility of sP-selectin as a biomarker of CVD in overweight/obese adults with schizophrenia (for example, by relating sP-selectin to incidence of cardiovascular events).
Collapse
Affiliation(s)
| | - Teodor T Postolache
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA, Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Denver Veterans Affairs Medical Center (VAMC), Aurora, CO, USA, Military and Veteran Microbiome Consortium for Research and Education (MVM-CoRE), Aurora, CO, USA, VISN 5 Capitol Health Care Network Mental Illness Research Education and Clinical Center (MIRECC), Baltimore, MD, USA
| | - Aline Dagdag
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dietmar Fuchs
- Division of Biological Chemistry, Biocenter Innsbruck Medical University, Innsbruck, Austria
| | - Olaoluwa O Okusaga
- Michael E. DeBakey VA Medical Center, Houston, TX, USA, Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
104
|
Abstract
The beneficial effects of a Mediterranean diet on human health and, in particular, on lowering risk of cardiovascular disease, has been mainly attributed to its high content to extra virgin olive oil (EVOO). While its main fatty acid, oleic acid, is considered important to these effects, EVOO has other biological properties that depend on, or are potentiated by other minor components of this oil. Initially, the mechanisms considered as possible causes of this cardioprotective effect of EVOO were based on the incidence on the so-called traditional risk factors (especially lipids and blood pressure). However, the high relative reduction in the prevalence of cardiovascular morbidity and mortality were not proportional to the limited findings about regulation of those traditional risk factors. In addition to several studies confirming the above effects, current research on beneficial effect of EVOO, and in particular in conjunction with Mediterranean style diets, is being focused on defining its effects on newer cardiovascular risk factors, such as inflammation, oxidative stress, coagulation, platelet aggregation, fibrinolysis, endothelial function or lipids or on the modulation of the conditions which predispose people to cardiovascular events, such as obesity, metabolic syndrome or type 2 diabetes mellitus. In the current review, we will mainly focus on reviewing the current evidence about the effects that EVOO exerts on alternative factors, including postprandial lipemia or coagulation, among others, discussing the underlying mechanism by which it exerts its effect, as well as providing a short review on future directions.
Collapse
|
105
|
Beurskens DMH, Huckriede JP, Schrijver R, Hemker HC, Reutelingsperger CP, Nicolaes GAF. The Anticoagulant and Nonanticoagulant Properties of Heparin. Thromb Haemost 2020; 120:1371-1383. [PMID: 32820487 DOI: 10.1055/s-0040-1715460] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Heparins represent one of the most frequently used pharmacotherapeutics. Discovered around 1926, routine clinical anticoagulant use of heparin was initiated only after the publication of several seminal papers in the early 1970s by the group of Kakkar. It was shown that heparin prevents venous thromboembolism and mortality from pulmonary embolism in patients after surgery. With the subsequent development of low-molecular-weight heparins and synthetic heparin derivatives, a family of related drugs was created that continues to prove its clinical value in thromboprophylaxis and in prevention of clotting in extracorporeal devices. Fundamental and applied research has revealed a complex pharmacodynamic profile of heparins that goes beyond its anticoagulant use. Recognition of the complex multifaceted beneficial effects of heparin underscores its therapeutic potential in various clinical situations. In this review we focus on the anticoagulant and nonanticoagulant activities of heparin and, where possible, discuss the underlying molecular mechanisms that explain the diversity of heparin's biological actions.
Collapse
Affiliation(s)
- Danielle M H Beurskens
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Joram P Huckriede
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Roy Schrijver
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - H Coenraad Hemker
- Synapse BV, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Chris P Reutelingsperger
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Gerry A F Nicolaes
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
106
|
Hounkpe BW, Benatti RDO, Carvalho BDS, De Paula EV. Identification of common and divergent gene expression signatures in patients with venous and arterial thrombosis using data from public repositories. PLoS One 2020; 15:e0235501. [PMID: 32780732 PMCID: PMC7418995 DOI: 10.1371/journal.pone.0235501] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 06/17/2020] [Indexed: 12/31/2022] Open
Abstract
STRENGTHS AND LIMITATIONS OF THIS STUDY Our results represent the first comparison of venous and arterial thrombosis at the transcriptomic level.Our main result was the demonstration that immunothrombosis pathways are important to the pathophysiology of these conditions, also at the transcriptomic level.A specific signature for venous and arterial thrombosis was described, and validated in independent cohorts.The limited number of public repositories with gene expression data from patients with venous thromboembolism limits the representation of these patients in our analyses.In order to gather a meaningful number of studies with gene expression data we had to include patients in different time-points since the index thrombotic event, which might have increased the heterogeneity of our population.
Collapse
Affiliation(s)
| | | | - Benilton de Sá Carvalho
- Department of Statistics, Institute of Mathematics, Statistics and Scientific Computing, University of Campinas, Campinas, SP, Brazil
| | - Erich Vinicius De Paula
- School of Medical Sciences, University of Campinas, Campinas, SP, Brazil
- Hematology and Hemotherapy Center, University of Campinas, Campinas, SP, Brazil
| |
Collapse
|
107
|
Papadaki S, Sidiropoulou S, Moschonas IC, Tselepis AD. Factor Xa and thrombin induce endothelial progenitor cell activation. The effect of direct oral anticoagulants. Platelets 2020; 32:807-814. [PMID: 32762584 DOI: 10.1080/09537104.2020.1802413] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Factor Xa (FXa) and thrombin exert non-hemostatic cellular actions primarily mediated through protease-activated receptors (PARs). We investigated the effect of FXa and thrombin on human late-outgrowth endothelial cells (OECs), a type of endothelial progenitor cells (EPCs), and on human umbilical vein endothelial cells (HUVECs). The effect of direct oral anticoagulants (DOACs), rivaroxaban and dabigatran, was also studied. The membrane expression of intercellular adhesion molecule-1 (ICAM-1) and the secretion of monocyte chemoattractant protein-1 (MCP-1) were used as cell activation markers. FXa and thrombin increase the ICAM-1 expression and the MCP-1 secretion on both cells, being higher on OECs. Vorapaxar, a specific PAR-1 antagonist, completely inhibits FXa-induced activation of both cells and thrombin-induced HUVEC activation, but only partially thrombin-induced OEC activation. Furthermore, thrombin-receptor activating peptide; TRAP-6, only partially activates OECs. OECs do not membrane-express PAR-4, therefore it may not be involved on thrombin-induced OEC activation. Rivaroxaban and dabigatran inhibit OEC and HUVEC activation by FXa and thrombin, respectively. Rivaroxaban enhances thrombin-induced OEC and HUVEC activation, which is completely inhibited by vorapaxar. The inhibition of OEC and HUVEC activation by vorapaxar and DOACs may represent a new pleiotropic effect of these drugs. The pathophysiological and clinical significance of our findings need to be established.
Collapse
Affiliation(s)
- Styliani Papadaki
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Sofia Sidiropoulou
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Iraklis C Moschonas
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Alexandros D Tselepis
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| |
Collapse
|
108
|
Correale M, Leopizzi A, Mallardi A, Ranieri A, Suriano MP, D'Alessandro D, Tricarico L, Mazzeo P, Tucci S, Pastore G, Maulucci G, Di Biase M, Brunetti ND. Switch to direct anticoagulants and improved endothelial function in patients with chronic heart failure and atrial fibrillation. Thromb Res 2020; 195:16-20. [PMID: 32634728 DOI: 10.1016/j.thromres.2020.06.046] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/07/2020] [Accepted: 06/26/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Chronic heart failure (CHF) is characterized by higher rates of atrial fibrillation (AF) and endothelial dysfunction (ED). First line anticoagulant therapy in AF is represented by direct oral anticoagulants (DOACs); several patients, however, are still treated with vitamin-K inhibitors. The use of DOACs is associated in previous studies with an improved vascular function. We therefore sought to evaluate possible changes in endothelial function assessed by flow-mediated dilation (FMD) in patients with CHF and AF shifting from warfarin to DOACs. METHODS Forty-three consecutive outpatients were enrolled in the study. FMD was assessed at baseline and after 4 months. Patients were compared according to AC therapy. RESULTS After the first measurement of FMD, 18 patients "switched" to DOACs because of poor compliance to warfarin therapy or time in therapeutic range, 19 patients continued to use DOACs, 6 warfarin. "Switched" patients to DOACs therapy showed an improved FMD (19.0 ± 6.6% vs 3.8 ± 1.3%, p < 0.0001); C-reactive protein (CRP) levels decreased in "switched" patients from 1.4 ± 0.5 to 1.0 ± 0.7 mg/dl (p < 0.05). FMD and CRP changes were not significant in patients who did not changed anticoagulant therapy. In switched patients, changes in CRP levels were proportional to FMD changes (r = -0.50, p < 0.05). Shifting from warfarin to DOACs was significantly correlated to improved FMD levels even at multivariable analysis (p < 0.05). CONCLUSIONS Switch from warfarin to DOACs in patents with CHF and AF was associated in an observational non randomized study with an improved endothelial function. Changes in FMD values were related to changes in CRP levels.
Collapse
Affiliation(s)
- Michele Correale
- Cardiology Department, Ospedali Riuniti University Hospital, Foggia, Italy
| | - Alessandra Leopizzi
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Adriana Mallardi
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Alessandro Ranieri
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Miriam Pia Suriano
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Damiano D'Alessandro
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Lucia Tricarico
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Pietro Mazzeo
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Salvatore Tucci
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Giuseppe Pastore
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy.
| | - Guglielmo Maulucci
- Cardiology Department, Ospedali Riuniti University Hospital, Foggia, Italy
| | - Matteo Di Biase
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | | |
Collapse
|
109
|
Döhrmann M, Makhoul S, Gross K, Krause M, Pillitteri D, von Auer C, Walter U, Lutz J, Volf I, Kehrel BE, Jurk K. CD36-fibrin interaction propagates FXI-dependent thrombin generation of human platelets. FASEB J 2020; 34:9337-9357. [PMID: 32463151 DOI: 10.1096/fj.201903189r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/11/2020] [Accepted: 04/28/2020] [Indexed: 12/27/2022]
Abstract
Thrombin converts fibrinogen to fibrin and activates blood and vascular cells in thrombo-inflammatory diseases. Platelets are amplifiers of thrombin formation when activated by leukocyte- and vascular cell-derived thrombin. CD36 on platelets acts as sensitizer for molecules with damage-associated molecular patterns, thereby increasing platelet reactivity. Here, we investigated the role of CD36 in thrombin-generation on human platelets, including selected patients with advanced chronic kidney disease (CKD). Platelets deficient in CD36 or blocked by anti-CD36 antibody FA6.152 showed impaired thrombin generation triggered by thrombin in calibrated automated thrombography. Using platelets with congenital function defects, blocking antibodies, pharmacological inhibitors, and factor-depleted plasma, CD36-sensitive thrombin generation was dependent on FXI, fibrin, and platelet signaling via GPIbα and SFKs. CD36-deficiency or blocking suppressed thrombin-induced platelet αIIbβ3 activation, granule exocytosis, binding of adhesion proteins and FV, FVIII, FIX, FX, but not anionic phospholipid exposure determined by flow cytometry. CD36 ligated specifically soluble fibrin, which recruited distinct coagulation factors via thiols. Selected patients with CKD showed elevated soluble fibrin plasma levels and enhanced thrombin-induced thrombin generation, which was normalized by CD36 blocking. Thus, CD36 is an important amplifier of platelet-dependent thrombin generation when exposure of anionic phospholipids is limited. This pathway might contribute to hypercoagulability in CKD.
Collapse
Affiliation(s)
- Mareike Döhrmann
- Center for Thrombosis and Hemostasis (CTH), University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Stephanie Makhoul
- Center for Thrombosis and Hemostasis (CTH), University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Kathrin Gross
- Center for Thrombosis and Hemostasis (CTH), University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Manuela Krause
- Deutsche Klinik für Diagnostik HELIOS Klinik, Wiesbaden, Germany
| | | | - Charis von Auer
- Third Department of Medicine, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Ulrich Walter
- Center for Thrombosis and Hemostasis (CTH), University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Jens Lutz
- Section of Nephrology, I. Department of Medicine, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany.,Medical Clinic, Section of Nephrology and Infectious Diseases, Gemeinschaftsklinikum Mittelrhein, Koblenz, Germany
| | - Ivo Volf
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Beate E Kehrel
- Department of Anesthesiology, Intensive Care and Pain Medicine, Experimental and Clinical Hemostasis, University of Muenster, Muenster, Germany
| | - Kerstin Jurk
- Center for Thrombosis and Hemostasis (CTH), University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany.,Department of Anesthesiology, Intensive Care and Pain Medicine, Experimental and Clinical Hemostasis, University of Muenster, Muenster, Germany
| |
Collapse
|
110
|
Golia N, Krishan K, Kashyap JR. Assessment of Obesity by Using Various Anthropometric Measurements among Patients with Coronary Heart Disease Residing in North India. Cureus 2020; 12:e7948. [PMID: 32509474 PMCID: PMC7270879 DOI: 10.7759/cureus.7948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Background Obesity is one of the main risk factors of coronary heart disease (CHD). Although a range of anthropometric measures are available to evaluate obesity, which measure is the most precise to predict the risk of CHD is still controversial. Therefore, we assess the prevalence of obesity among patients with CHD by using various anthropometric techniques to find out the most efficient method to predict the risk of CHD. Methods In this cross-sectional study, we included 300 CHD patients and 100 age and sex-matched healthy controls, aged 45-70 years. Various anthropometric measurements (waist and hip circumference, waist-hip ratio, body mass index, and body fat percentage) were taken to assess the prevalence of obesity among the selected population. Results Average waist circumference among male and female patients was significantly higher than the controls; 94.0±13.2 vs. 86.4±4.4 (p < 0.001) and 97.8±12.1 vs. 86.9±5.3 (p < 0.001) respectively. The average waist-hip ratio among patients of both genders was significantly higher than controls 1.0±.06 vs. 0.92±.04 (p < 0.001) among males and 0.96±.07 vs. 0.88±.04 (p < 0.001) among females, respectively. The average body mass index (BMI) was not different among male patients (24.6±4.0) compared to controls (24.3±2.3); however, the frequency distribution of BMI among male patients and controls was significantly different (p < 0.05), whereas female patients had significantly higher BMI compared to controls 27.7±4.9 and 25.1±2.4, (p < 0.001). Similarly, body fat percentage and visceral fat percentage were elevated among female cases vs. female controls, but no significant difference was observed in the body fat percentage of male cases vs. controls 28.0±5.0 vs. 28.1±2.7; (p > 0.05). However, visceral fat percentage was significantly elevated among male cases vs. controls 11.6±5.7 vs. 9.6±2.6 (p < 0.05). Conclusions Central adiposity markers, waist circumference (WC), waist-hip ratio (WHR), and visceral fat percentage were uniformly present in patients of both sexes and are stronger predictors of risk of CHD relative to the BMI.
Collapse
Affiliation(s)
- Nisha Golia
- Anthropology, Panjab University, Chandigarh, IND
| | | | - Jeet Ram Kashyap
- Cardiology, Government Medical College and Hospital Sector, Chandigarh, IND
| |
Collapse
|
111
|
Lordan R, Tsoupras A, Zabetakis I. Platelet activation and prothrombotic mediators at the nexus of inflammation and atherosclerosis: Potential role of antiplatelet agents. Blood Rev 2020; 45:100694. [PMID: 32340775 DOI: 10.1016/j.blre.2020.100694] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 03/22/2020] [Accepted: 04/07/2020] [Indexed: 12/20/2022]
Abstract
Platelets are central to inflammation-related manifestations of cardiovascular diseases (CVD) such as atherosclerosis. Platelet-activating factor (PAF), thrombin, thromboxane A2 (TxA2), and adenosine diphosphate (ADP) are some of the key agonists of platelet activation that are at the intersection between a plethora of inflammatory pathways that modulate pro-inflammatory and coagulation processes. The aim of this article is to review the role of platelets and the relationship between their structure, function, and the interactions of their constituents in systemic inflammation and atherosclerosis. Antiplatelet therapies are discussed with a view to primary prevention of CVD by the clinical reduction of platelet reactivity and inflammation. Current antiplatelet therapies are effective in reducing cardiovascular risk but increase bleeding risk. Novel therapeutic antiplatelet approaches beyond current pharmacological modalities that do not increase the risk of bleeding require further investigation. There is potential for specifically designed nutraceuticals that may become safer alternatives to pharmacological antiplatelet agents for the primary prevention of CVD but there is serious concern over their efficacy and regulation, which requires considerably more research.
Collapse
Affiliation(s)
- Ronan Lordan
- Department of Biological Sciences, University of Limerick, Limerick, Ireland; Health Research Institute (HRI), University of Limerick, Limerick, Ireland; Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-5158, USA.
| | - Alexandros Tsoupras
- Department of Biological Sciences, University of Limerick, Limerick, Ireland; Health Research Institute (HRI), University of Limerick, Limerick, Ireland
| | - Ioannis Zabetakis
- Department of Biological Sciences, University of Limerick, Limerick, Ireland; Health Research Institute (HRI), University of Limerick, Limerick, Ireland
| |
Collapse
|
112
|
Chen Z, Li N, Wang J, Li C, He S, Zhou X, He Y. Association between mean platelet volume and major adverse cardiac events in percutaneous coronary interventions: a systematic review and meta-analysis. Coron Artery Dis 2020; 31:722-732. [DOI: 10.1097/mca.0000000000000885] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
113
|
Xu JH, Lu SJ, Wu P, Kong LC, Ning C, Li HY. Molecular mechanism whereby paraoxonase-2 regulates coagulation activation through endothelial tissue factor in rat haemorrhagic shock model. Int Wound J 2020; 17:735-741. [PMID: 32090497 DOI: 10.1111/iwj.13329] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/05/2020] [Accepted: 02/09/2020] [Indexed: 01/17/2023] Open
Abstract
We investigated the molecular mechanism of paraoxonase-2 (PON-2) in regulating blood coagulation activation in rats with haemorrhagic shock through endothelial tissue factor (TF). Thirty adult Sprague Dawley rats were randomly divided into three groups: healthy control group (group A), the haemorrhagic shock PON-2 treatment group (group B), and the haemorrhagic shock group (group C). After the model was established, blood was withdrawn from the inferior vena cava of all rats. The difference in plasma thrombomodulin (TM) levels of the three groups was determined by Western blotting. The expression of transcription factors Egr-1 and Sp1 was detected by Western blotting assays. reverse transcription-polymerase chain Reaction (RT-PCR) was used to determine the mRNA expression of t-PA, PAI-1, TM, and PON-2 in the serum of three groups of rats. Endothelial TF was measured by enzyme linked immunosorbent assay (ELISA), and coagulation assay was used to detect the activity of coagulation factor VIII. Histopathological examination of the arteries of the rats was performed. The molecular mechanism of PON-2 in regulating blood coagulation activation in haemorrhagic shock model rats by endothelial tissue factor was analysed. The expression of thrombin was determined by electrophoresis. Compared with the healthy control group, the expression of TM in groups B and C decreased, both 188.64 ± 12.47 and 137.48 ± 9.72, respectively, with a significant difference. The mRNA expression of TM and PON was determined by RT-PCR. The mRNA expression of TM and PON in group B was 0.97 ± 0.07 and 1.14 ± 0.09, compared with the control group, and the mRNA expression of TM and PON in group C was 0.86 ± 0.38 and 1.12 ± 0.41, both of which increased, and there were significant differences. By measuring the expression of endothelial TF, the expression of TF in groups B and C was elevated to 12.69 ± 1.07 and 11.59 ± 0.87, with significant differences. The enzyme activities of PON-2 in groups B and C, which were 110.34 ± 14.37 and 52.37 ± 8.06, respectively, were increased compared with the healthy control group and there were significant differences. PON-2 regulates the activation of coagulation in rats with haemorrhagic shock by regulating the expression of endothelial tissue-related genes such as plasma TM and endothelial TF under hypoxic and ischaemic conditions.
Collapse
Affiliation(s)
- Jian-Hua Xu
- Intensive Care Unit, Linyi Central Hospital, Linyi, Shandong, China
| | - Shi-Jun Lu
- Intensive Care Unit, Linyi Central Hospital, Linyi, Shandong, China
| | - Peng Wu
- Intensive Care Unit, Linyi Central Hospital, Linyi, Shandong, China
| | - Ling-Chen Kong
- Intensive Care Unit, Linyi Central Hospital, Linyi, Shandong, China
| | - Chao Ning
- Intensive Care Unit, Linyi Central Hospital, Linyi, Shandong, China
| | - Hai-Yan Li
- Intensive Care Unit, Linyi Central Hospital, Linyi, Shandong, China
| |
Collapse
|
114
|
Gurbel PA, Fox KAA, Tantry US, Ten Cate H, Weitz JI. Combination Antiplatelet and Oral Anticoagulant Therapy in Patients With Coronary and Peripheral Artery Disease. Circulation 2020; 139:2170-2185. [PMID: 31034291 DOI: 10.1161/circulationaha.118.033580] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Antiplatelet therapy is the mainstay for the treatment of acute and chronic arterial disease involving the coronary and peripheral beds. However, questions remain about optimal antithrombotic therapy for long-term treatment of chronic vascular disease. The observation that dual antiplatelet therapy with acetylsalicylic acid and clopidogrel was associated with lower thrombotic event rates than acetylsalicylic acid monotherapy in patients with acute coronary syndromes and those undergoing percutaneous coronary intervention changed the treatment paradigm. Moreover, the demonstration that more pharmacodynamically potent P2Y12 inhibitors than clopidogrel were associated with fewer thrombotic event occurrences further solidified the dual antiplatelet therapy approach. However, recurrent thrombotic events occur in ≈1 in 10 patients in the first year following an acute coronary syndrome event, despite treatment with the most potent P2Y12 inhibitors, a limitation that has stimulated interest in exploring the efficacy and safety of approaches using anticoagulants on top of antiplatelet therapy. These investigations have included treatment with very-low-dose oral anticoagulation, and even its replacement of acetylsalicylic acid in the presence of a P2Y12 inhibitor, in patients stabilized after an acute coronary syndrome event. Recent basic and translational studies have suggested noncanonical effects of coagulation factor inhibition that may further modulate clinical benefits. This in-depth review will discuss developments in our understanding of the roles that platelets and coagulation factors play in atherothrombosis and review the rationale and clinical evidence for combining antiplatelet and oral anticoagulant therapy in patients with coronary and peripheral artery disease.
Collapse
Affiliation(s)
- Paul A Gurbel
- Inova Center for Thrombosis Research and Drug Development, Inova Heart and Vascular Institute, Falls Church, VA (P.A.G.)
| | - Keith A A Fox
- British Heart Foundation Centre for Cardiovascular Sciences, University of Edinburgh, United Kingdom (K.A.A.F.)
| | - Udaya S Tantry
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, MD (U.S.T.)
| | - Hugo Ten Cate
- Laboratory for Clinical Thrombosis and Hemostasis, Departments of Internal Medicine and Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Netherlands (H.t.C.)
| | - Jeffrey I Weitz
- Thrombosis and Atherosclerosis Research Institute and McMaster University, Hamilton, Canada (J.I.W.)
| |
Collapse
|
115
|
Morsy MD. Hemostatic effect of acylated ghrelin in control and sleeve gastrectomy-induced rats: mechanisms of action. Arch Physiol Biochem 2020; 126:31-40. [PMID: 30320517 DOI: 10.1080/13813455.2018.1489849] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
This study investigated the effect of acylated ghrelin (AG) deficiency after sleeve gastrectomy (SG) or chronic administration in control and SG-indiuced rats on platelet function, coagulation, and fibrinolysis. Administration of AG (100 µg/kg, subcutaneously) to control or SG rats significantly inhibited platelets aggregation and lowered levels of Von-Willebrand factor (vWF), fibrinogen, and thromboxane B2. Concomitantly, it decreased circulatory levels and aortic expression levels of plasminogen activator inhibitor-1 (PAI-1) and tissue factor (TF) and increased the aortic expression of the endothelial nitric oxidase (eNOS). However, AG inhibited angiotensin-II (ANGII)-induced upregulation of tissue factor pathway inhibitor (TPAI) and TF and increased activity of TF and increases eNOS expression in cultured endothelial cells, an effect that was abolished by the addition of D-[lys3]-GHRP-6, a selective AG receptor (GHSR-1a) blocker or L-Name, a potent eNOS inhibitor. In conclusion, AG has an anti-platelet, anti-coagulant, and fibrinolytic roles mediated through GHSR-1a to enhance nitric oxide synthesis.
Collapse
Affiliation(s)
- Mohamed Darwesh Morsy
- Department of Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
- Department of Physiology, College of Medicine, Menoufia University, Shebeen Alkoom, Egypt
| |
Collapse
|
116
|
Sychev DA, Baturina OA, Mirzaev KB, Rytkin E, Ivashchenko DV, Andreev DA, Ryzhikova KA, Grishina EA, Bochkov PO, Shevchenko RV. CYP2C19*17 May Increase the Risk of Death Among Patients with an Acute Coronary Syndrome and Non-Valvular Atrial Fibrillation Who Receive Clopidogrel and Rivaroxaban. Pharmgenomics Pers Med 2020; 13:29-37. [PMID: 32158254 PMCID: PMC6986167 DOI: 10.2147/pgpm.s234910] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 01/03/2020] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION The aim of this study is to assess the influence of gene CYP2C19, CYP3A4, CYP3A5 and ABCB1 polymorphisms on clopidogrel antiplatelet activity, rivaroxaban concentration equilibrium, and clinical outcomes among patients with acute coronary syndrome and non-valvular atrial fibrillation. METHODS In the multicenter prospective registry study of the efficacy and safety of a combined antithrombotic therapy 103 patients with non-valvular atrial fibrillation both undergoing or not a percutaneous coronary intervention were enrolled. The trial assessed the primary outcomes (major bleeding, in-hospital death, cardiovascular death, stroke\transient ischaemic attack, death/renal insufficiency) and secondary outcomes (platelet reactivity units (PRU), rivaroxaban concentration). RESULTS For none of the clinical outcomes when combined with other covariates, the carriership of polymorphisms CYP3A5*3 rs776746, CYP2C19*2 rs4244285;*17 rs12248560, ABCB1 3435 C>T, ABCB1 rs4148738 was significant. None of the markers under study (CYP3A5*3 rs776746, CYP2C19*2 rs4244285, *17 rs12248560, ABCB1 3435 C>T, ABCB1 rs4148738) has proven to affect rivaroxaban equilibrium concentration in blood plasma among patients with atrial fibrillation and acute coronary syndrome. CONCLUSION In situations of double or triple antithrombotic rivaroxaban and clopidogrel therapy among patients with atrial fibrillation and acute coronary syndrome, the genetic factors associated with bleeding complications risk (CYP2C19*17) may prove to be clinically relevant.
Collapse
Affiliation(s)
- D A Sychev
- Russian Medical Academy of Continuous Professional Education, Ministry of Health of the Russian Federation, Moscow, Russian Federation
| | | | - K B Mirzaev
- Russian Medical Academy of Continuous Professional Education, Ministry of Health of the Russian Federation, Moscow, Russian Federation
| | - E Rytkin
- Russian Medical Academy of Continuous Professional Education, Ministry of Health of the Russian Federation, Moscow, Russian Federation
| | - D V Ivashchenko
- Russian Medical Academy of Continuous Professional Education, Ministry of Health of the Russian Federation, Moscow, Russian Federation
| | - D A Andreev
- Sechenov University, Moscow, Russian Federation
| | - K A Ryzhikova
- Russian Medical Academy of Continuous Professional Education, Ministry of Health of the Russian Federation, Moscow, Russian Federation
| | - E A Grishina
- Russian Medical Academy of Continuous Professional Education, Ministry of Health of the Russian Federation, Moscow, Russian Federation
| | - P O Bochkov
- Russian Medical Academy of Continuous Professional Education, Ministry of Health of the Russian Federation, Moscow, Russian Federation
| | - R V Shevchenko
- Russian Medical Academy of Continuous Professional Education, Ministry of Health of the Russian Federation, Moscow, Russian Federation
| |
Collapse
|
117
|
Capodanno D, Bhatt DL, Eikelboom JW, Fox KAA, Geisler T, Michael Gibson C, Gonzalez-Juanatey JR, James S, Lopes RD, Mehran R, Montalescot G, Patel M, Steg PG, Storey RF, Vranckx P, Weitz JI, Welsh R, Zeymer U, Angiolillo DJ. Dual-pathway inhibition for secondary and tertiary antithrombotic prevention in cardiovascular disease. Nat Rev Cardiol 2020; 17:242-257. [PMID: 31953535 DOI: 10.1038/s41569-019-0314-y] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/18/2019] [Indexed: 12/24/2022]
Abstract
Advances in antiplatelet therapies for patients with cardiovascular disease have improved patient outcomes over time, but the challenge of balancing the risks of ischaemia and bleeding remains substantial. Moreover, many patients with cardiovascular disease have a residual risk of ischaemic events despite receiving antiplatelet therapy. Therefore, novel strategies are needed to prevent clinical events through mechanisms beyond platelet inhibition and with an acceptable associated risk of bleeding. The advent of non-vitamin K antagonist oral anticoagulants, which attenuate fibrin formation by selective inhibition of factor Xa or thrombin, has renewed the interest in dual-pathway inhibition strategies that combine an antiplatelet agent with an anticoagulant drug. In this Review, we highlight the emerging pharmacological rationale and clinical development of dual-pathway inhibition strategies for the prevention of atherothrombotic events in patients with different manifestations of cardiovascular disease, such as coronary artery disease, cerebrovascular disease and peripheral artery disease.
Collapse
Affiliation(s)
- Davide Capodanno
- Division of Cardiology, C.A.S.T., P.O. 'G. Rodolico', Azienda Ospedaliero-Universitaria 'Policlinico-Vittorio Emanuele', University of Catania, Catania, Italy
| | - Deepak L Bhatt
- Brigham and Women's Hospital Heart and Vascular Center, Harvard Medical School, Boston, MA, USA
| | - John W Eikelboom
- Population Health Research Institute, Hamilton General Hospital and McMaster University, Hamilton, ON, Canada
| | - Keith A A Fox
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Tobias Geisler
- Department of Cardiology and Angiology, University Hospital of Tübingen, Tübingen, Germany
| | - C Michael Gibson
- Cardiovascular Division, Department of Medicine, Beth Israel Deaconess Medical, Harvard Medical School, Boston, MA, USA
| | | | - Stefan James
- Department of Medical Sciences and Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Renato D Lopes
- Division of Cardiology, Duke Clinical Research Institute, Duke University, Durham, NC, USA
| | - Roxana Mehran
- Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gilles Montalescot
- Sorbonne Université, ACTION Study Group, Institut de Cardiologie, Pitié Salpêtrière Hôpital (AP-HP), Paris, France
| | - Manesh Patel
- Division of Cardiology, Duke Clinical Research Institute, Duke University, Durham, NC, USA
| | - P Gabriel Steg
- Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, Paris University, FACT (French Alliance for Cardiovascular Trials), INSERM U1148, Paris, France
| | - Robert F Storey
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Pascal Vranckx
- Department of Cardiology and Critical Care Medicine, Hartcentrum Hasselt, and Faculty of Medicine and Life Sciences at the University of Hasselt, Hasselt, Belgium
| | - Jeffrey I Weitz
- Departments of Medicine and Biochemistry and Biomedical Sciences, McMaster University and the Thrombosis and Atherosclerosis Research Institute, Hamilton, ON, Canada
| | - Robert Welsh
- Cardiac Sciences Department, Mazankowski Alberta Heart Institute and University of Alberta, Edmonton, AL, Canada
| | - Uwe Zeymer
- Klinikum Ludwigshafen and Institut für Herzinfarktforschung, Ludwigshafen, Germany
| | - Dominick J Angiolillo
- Division of Cardiology, University of Florida College of Medicine, Jacksonville, FL, USA.
| |
Collapse
|
118
|
Olivieri O, Turcato G, Moruzzi S, Castagna A, Girelli D, Pizzolo F, Friso S, Sandri M, Bassi A, Martinelli N. Not Just Arterial Damage: Increased Incidence of Venous Thromboembolic Events in Cardiovascular Patients With Elevated Plasma Levels of Apolipoprotein CIII. J Am Heart Assoc 2020; 8:e010973. [PMID: 30646800 PMCID: PMC6497332 DOI: 10.1161/jaha.118.010973] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Background Apolipoprotein CIII (apo CIII) is a crucial player in triglyceride‐rich lipoprotein metabolism, but may also act pleiotropically, provoking inflammatory responses and stimulating coagulation. Elevated apoCIII plasma levels have been associated with increased activity of coagulation factors. Since these features of prothrombotic diathesis are linked with venous thromboembolism (VTE), we hypothesized that apo CIII plays a role in VTE. Methods and Results We recorded nonfatal VTE events in 1020 patients (age 63.3±11.4 years; 29.1% women) with or without coronary artery disease (79.1% with coronary artery disease and 20.9% without coronary artery disease) during a long follow‐up. Complete plasma lipid and apolipoproteins were available for all patients. Forty‐five patients (4.4%) experienced nonfatal VTE events during a median follow‐up period of 144 months. Apo CIII plasma concentration at enrollment was higher in patients with VTE compared with patients without VTE (12.2 [95% CI, 11.10–13.5] mg/dL vs 10.6 [95% CI, 10.4–10.9] mg/dL, respectively; P=0.011). Patients with apo CIII levels above the median value (10.6 mg/dL) exhibited an increased risk of VTE (incidence rate, 6.0 [95% CI, 4.0–8.0] vs 1.8 [95% CI, 0.7–2.9] VTE events/1000 person‐years; unadjusted hazard ratio [HR], 3.42 [95% CI, 1.73–6.75]; P<0.001). This association was confirmed after adjustment for sex, age, coronary artery disease diagnosis, body mass index, hypertension, and anticoagulant treatment at enrollment (HR, 2.66; 95% CI, 1.31–5.37 [P=0.007]), with inclusion of lipid parameters in the Cox model (HR, 3.74; 95% CI, 1.24–11.33 [P=0.019]), and even with exclusion of patients who died at follow‐up (HR, 3.92; 95% CI, 1.68–9.14 [P=0.002]) or patients taking anticoagulants (HR, 3.39; 95% CI, 1.72–6.69 [P<0.001]). Conclusions Our results suggest that high plasma apo CIII concentrations may predict an increased risk of VTE in patients with cardiovascular disease.
Collapse
Affiliation(s)
- Oliviero Olivieri
- 1 Unit of Internal Medicine Department of Medicine University of Verona Italy
| | | | | | - Annalisa Castagna
- 1 Unit of Internal Medicine Department of Medicine University of Verona Italy
| | - Domenico Girelli
- 1 Unit of Internal Medicine Department of Medicine University of Verona Italy
| | - Francesca Pizzolo
- 1 Unit of Internal Medicine Department of Medicine University of Verona Italy
| | - Simonetta Friso
- 1 Unit of Internal Medicine Department of Medicine University of Verona Italy
| | - Marco Sandri
- 1 Unit of Internal Medicine Department of Medicine University of Verona Italy
| | - Antonella Bassi
- 1 Unit of Internal Medicine Department of Medicine University of Verona Italy.,3 Laboratory of Clinical Chemistry and Hematology University Hospital of Verona Italy
| | - Nicola Martinelli
- 1 Unit of Internal Medicine Department of Medicine University of Verona Italy
| |
Collapse
|
119
|
Olie RH, van der Meijden PEJ, Spronk HMH, Ten Cate H. Antithrombotic Therapy: Prevention and Treatment of Atherosclerosis and Atherothrombosis. Handb Exp Pharmacol 2020; 270:103-130. [PMID: 32776281 DOI: 10.1007/164_2020_357] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Atherosclerosis is a multifactorial vascular disease that develops in the course of a lifetime. Numerous risk factors for atherosclerosis have been identified, mostly inflicting pro-inflammatory effects. Vessel injury, such as occurring during erosion or rupture of atherosclerotic lesions triggers blood coagulation, in attempt to maintain hemostasis (protect against bleeding). However, thrombo-inflammatory mechanisms may drive blood coagulation such that thrombosis develops, the key process underlying myocardial infarction and ischemic stroke (not due to embolization from the heart). In the blood coagulation system, platelets and coagulation proteins are both essential elements. Hyperreactivity of blood coagulation aggravates atherosclerosis in preclinical models. Pharmacologic inhibition of blood coagulation, either with platelet inhibitors, or better documented with anticoagulants, or both, limits the risk of thrombosis and may potentially reverse atherosclerosis burden, although the latter evidence is still based on animal experimentation.Patients at risk of atherothrombotic complications should receive a single antiplatelet agent (acetylsalicylic acid, ASA, or clopidogrel); those who survived an atherothrombotic event will be prescribed temporary dual antiplatelet therapy (ASA plus a P2Y12 inhibitor) in case of myocardial infarction (6-12 months), or stroke (<6 weeks), followed by a single antiplatelet agent indefinitely. High risk for thrombosis patients (such as those with peripheral artery disease) benefit from a combination of an anticoagulant and ASA. The price of gained efficacy is always increased risk of (major) bleeding; while tailoring therapy to individual needs may limit the risks to some extent, new generations of agents that target less critical elements of hemostasis and coagulation mechanisms are needed to maintain efficacy while reducing bleeding risks.
Collapse
Affiliation(s)
- R H Olie
- Internal Medicine and CARIM School for Cardiovascular Research, Maastricht University Medical Center, Maastricht, The Netherlands.,Thrombosis Expertise Center, Heart+ Cardiovascular Center, and Department of Biochemistry, Maastricht University Medical Center, Maastricht, The Netherlands
| | - P E J van der Meijden
- Thrombosis Expertise Center, Heart+ Cardiovascular Center, and Department of Biochemistry, Maastricht University Medical Center, Maastricht, The Netherlands
| | - H M H Spronk
- Thrombosis Expertise Center, Heart+ Cardiovascular Center, and Department of Biochemistry, Maastricht University Medical Center, Maastricht, The Netherlands
| | - H Ten Cate
- Internal Medicine and CARIM School for Cardiovascular Research, Maastricht University Medical Center, Maastricht, The Netherlands. .,Thrombosis Expertise Center, Heart+ Cardiovascular Center, and Department of Biochemistry, Maastricht University Medical Center, Maastricht, The Netherlands.
| |
Collapse
|
120
|
Di Fusco SA, Lucà F, Gulizia MM, Gabrielli D, Colivicchi F. Emerging clinical setting of direct oral anticoagulants. J Cardiovasc Med (Hagerstown) 2020; 21:1-5. [DOI: 10.2459/jcm.0000000000000894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
121
|
Sarioglu O, Capar AE, Belet U. Relationship of arteriovenous fistula stenosis and thrombosis with the platelet-lymphocyte ratio in hemodialysis patients. J Vasc Access 2019; 21:630-635. [PMID: 31884875 DOI: 10.1177/1129729819894113] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The platelet-lymphocyte ratio, which was reported to have a strong relationship with chronic inflammation and thrombosis, is a useful biomarker. The purpose of this study was to evaluate the relationship between the platelet-lymphocyte ratio, arteriovenous stenosis, and thrombosis in patients with chronic renal failure. METHODS Patients who were referred to our interventional radiology department due to arteriovenous fistula dysfunction from dialysis units between August 2015 and December 2018 were retrospectively reviewed. In the study, 95 patients with arteriovenous fistula access problems were included. Patients were divided into two groups: stenosis (n = 52) and thrombosis (n = 43). Thirty-six subjects with a patent left radiocephalic arteriovenous fistula proven by both color Doppler ultrasonography and clinically were added to the control group. Blood samples were obtained on the same day before the fistulography. RESULTS Platelet counts, lymphocyte counts, and platelet-lymphocyte ratio were found to be significantly different between the three groups. After the Bonferroni post hoc analysis, there was a significant difference between the stenosis and control group (p = 0.017), and the thrombosis and control group (p < 0.001) in terms of the platelet-lymphocyte ratio. No significant difference for any parameter was found between stenosis and thrombosis group. CONCLUSION High levels of the platelet-lymphocyte ratio may be a supportive finding of arteriovenous fistula stenosis and thrombosis and can be taken into consideration during hemodialysis-dependent patients' follow-up.
Collapse
Affiliation(s)
- Orkun Sarioglu
- Department of Radiology, Izmir University of Health Sciences Tepecik Training and Research Hospital, Izmir, Turkey
| | - Ahmet Ergin Capar
- Department of Radiology, Izmir University of Health Sciences Tepecik Training and Research Hospital, Izmir, Turkey
| | - Umit Belet
- Department of Radiology, Izmir University of Health Sciences Tepecik Training and Research Hospital, Izmir, Turkey
| |
Collapse
|
122
|
Coppini R, Santini L, Palandri C, Sartiani L, Cerbai E, Raimondi L. Pharmacological Inhibition of Serine Proteases to Reduce Cardiac Inflammation and Fibrosis in Atrial Fibrillation. Front Pharmacol 2019; 10:1420. [PMID: 31956307 PMCID: PMC6951407 DOI: 10.3389/fphar.2019.01420] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 11/07/2019] [Indexed: 12/18/2022] Open
Abstract
Systemic inflammation correlates with an increased risk of atrial fibrillation (AF) and thrombogenesis. Systemic inflammation alters vessel permeability, allowing inflammatory and immune cell migration toward target organs, including the heart. Among inflammatory cells infiltrating the atria, macrophages and mast cell have recently attracted the interest of basic researchers due to the pathogenic mechanisms triggered by their activation. This chemotactic invasion is likely implicated in short- and long-term changes in cardiac cell-to-cell communication and in triggering fibrous tissue accumulation in the atrial myocardium and electrophysiological re-arrangements of atrial cardiomyocytes, thus favoring the onset and progression of AF. Serine proteases are a large and heterogeneous class of proteases involved in several processes that are important for cardiac function and are involved in cardiac diseases, such as (i) coagulation, (ii) fibrinolysis, (iii) extracellular matrix degradation, (iv) activation of receptors (i.e., protease-activated receptors [PPARs]), and (v) modulation of the activity of endogenous signals. The recognition of serine proteases substrates and their involvement in inflammatory/profibrotic mechanisms allowed the identification of novel cardio-protective mechanisms for commonly used drugs that inhibit serine proteases. The aim of this review is to summarize knowledge on the role of inflammation and fibrosis as determinants of AF. Moreover, we will recapitulate current findings on the role of serine proteases in the pathogenesis of AF and the possible beneficial effects of drugs inhibiting serine proteases in reducing the risk of AF through decrease of cardiac inflammation and fibrosis. These drugs include thrombin and factor Xa inhibitors (used as oral anticoagulants), dipeptidyl-peptidase 4 (DPP4) inhibitors, used for type-2 diabetes, as well as novel experimental inhibitors of mast cell chymases.
Collapse
Affiliation(s)
- Raffaele Coppini
- Section of Pharmacology, Department of Neurology, Psychology, Drug Sciences and Child Health, University of Florence, Florence, Italy
| | - Lorenzo Santini
- Section of Pharmacology, Department of Neurology, Psychology, Drug Sciences and Child Health, University of Florence, Florence, Italy
| | - Chiara Palandri
- Section of Pharmacology, Department of Neurology, Psychology, Drug Sciences and Child Health, University of Florence, Florence, Italy
| | - Laura Sartiani
- Section of Pharmacology, Department of Neurology, Psychology, Drug Sciences and Child Health, University of Florence, Florence, Italy
| | - Elisabetta Cerbai
- Section of Pharmacology, Department of Neurology, Psychology, Drug Sciences and Child Health, University of Florence, Florence, Italy
| | - Laura Raimondi
- Section of Pharmacology, Department of Neurology, Psychology, Drug Sciences and Child Health, University of Florence, Florence, Italy
| |
Collapse
|
123
|
Biedermann JS, Kruip MJHA, van der Meer FJ, Rosendaal FR, Leebeek FWG, Cannegieter SC, Lijfering WM. Rosuvastatin use improves measures of coagulation in patients with venous thrombosis. Eur Heart J 2019; 39:1740-1747. [PMID: 29394348 DOI: 10.1093/eurheartj/ehy014] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Accepted: 01/11/2018] [Indexed: 12/22/2022] Open
Abstract
Aims Observational studies indicate that statins reduce the risk of recurrent venous thrombosis (VT). However, trials have not been performed and the mechanism is unknown. We aimed to determine whether statin therapy improves the coagulation profile in patients with prior VT. Methods and results Randomized clinical trial (NCT01613794). Patients were randomized to rosuvastatin 20 mg/day for 4 weeks or no intervention. Blood was drawn at baseline and at end of study. The primary outcome was factor (F) VIII:C. In total, five coagulation factors were measured: FVIII:C, von Willebrand factor:Ag, FVII:C, FXI:C, and D-dimer. Among 247 randomized participants, mean age was 58 years, 62% were women and 49% had unprovoked VT. For all tested coagulation factors, mean levels were clearly decreased at end of study in rosuvastatin users, whereas they hardly differed in non-statin users. Results were most consistent for FVIII:C where mean FVIII:C levels were 7.2 IU/dL [95% CI (confidence interval) 2.9-11.5] lower in rosuvastatin users, while among non-users, no change in FVIII:C was observed (mean difference -0.1; 95% CI -3.0 to 2.9). The mean age and sex adjusted difference in FVIII:C change was -6.7 IU/dL (95% CI -12.0 to -1.4) in rosuvastatin users vs. non-users. Subgroup analyses revealed that the decrease in coagulation factors by rosuvastatin was more pronounced in participants with unprovoked VT and in those with cardiovascular risk factors. Conclusion Rosuvastatin 20 mg/day substantially improved the coagulation profile among patients with prior VT. These results suggest that statin therapy might be beneficial in patients at risk of recurrent VT.
Collapse
Affiliation(s)
- Joseph S Biedermann
- Department of Hematology, Erasmus University Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.,Star-Medical Anticoagulation Clinic, Vlambloem 21, 3068 JE Rotterdam, The Netherlands
| | - Marieke J H A Kruip
- Department of Hematology, Erasmus University Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.,Star-Medical Anticoagulation Clinic, Vlambloem 21, 3068 JE Rotterdam, The Netherlands
| | - Felix J van der Meer
- Department of Thrombosis and Haemostasis, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Frits R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Frank W G Leebeek
- Department of Hematology, Erasmus University Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands
| | - Suzanne C Cannegieter
- Department of Thrombosis and Haemostasis, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands.,Department of Clinical Epidemiology, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Willem M Lijfering
- Department of Clinical Epidemiology, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| |
Collapse
|
124
|
Jaminon A, Reesink K, Kroon A, Schurgers L. The Role of Vascular Smooth Muscle Cells in Arterial Remodeling: Focus on Calcification-Related Processes. Int J Mol Sci 2019; 20:E5694. [PMID: 31739395 PMCID: PMC6888164 DOI: 10.3390/ijms20225694] [Citation(s) in RCA: 182] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 10/31/2019] [Accepted: 11/08/2019] [Indexed: 12/22/2022] Open
Abstract
Arterial remodeling refers to the structural and functional changes of the vessel wall that occur in response to disease, injury, or aging. Vascular smooth muscle cells (VSMC) play a pivotal role in regulating the remodeling processes of the vessel wall. Phenotypic switching of VSMC involves oxidative stress-induced extracellular vesicle release, driving calcification processes. The VSMC phenotype is relevant to plaque initiation, development and stability, whereas, in the media, the VSMC phenotype is important in maintaining tissue elasticity, wall stress homeostasis and vessel stiffness. Clinically, assessment of arterial remodeling is a challenge; particularly distinguishing intimal and medial involvement, and their contributions to vessel wall remodeling. The limitations pertain to imaging resolution and sensitivity, so methodological development is focused on improving those. Moreover, the integration of data across the microscopic (i.e., cell-tissue) and macroscopic (i.e., vessel-system) scale for correct interpretation is innately challenging, because of the multiple biophysical and biochemical factors involved. In the present review, we describe the arterial remodeling processes that govern arterial stiffening, atherosclerosis and calcification, with a particular focus on VSMC phenotypic switching. Additionally, we review clinically applicable methodologies to assess arterial remodeling and the latest developments in these, seeking to unravel the ubiquitous corroborator of vascular pathology that calcification appears to be.
Collapse
Affiliation(s)
- Armand Jaminon
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Koen Reesink
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Abraham Kroon
- Department of Internal Medicine, Maastricht University Medical Centre (MUMC+), 6229 HX Maastricht, The Netherlands;
| | - Leon Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| |
Collapse
|
125
|
Shatoor AS, Shati A, Al Humayed SM, Shatoor AK, Alhawiti NM, Alqahtani SA. The hypocoagulant effect of Crataegus aronia in rats entails vitamin K-dependent and vitamin K-independent effects. J Food Biochem 2019; 44:e13094. [PMID: 31702060 DOI: 10.1111/jfbc.13094] [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] [Received: 07/11/2019] [Revised: 10/11/2019] [Accepted: 10/14/2019] [Indexed: 01/10/2023]
Abstract
This study investigated the effect of the aqueous extract of Crataegus aronia on blood coagulation in rats. Rats (200 ± 10 g,) were divided into two groups (6 rats/each) of control or C. aronia-treated rats which treated with the vehicle or the extract (200 mg/kg) for 21 days. With normal liver structure, serum levels of ALT, AST and ɣ-GT, platelet count, and plasma levels of vWF, values of PT and aPTT were significantly increased in C. aronia-treated rats. Also, it lowered serum levels of vitamin K (VK) and plasma activities of FII, FV, FVII, FVIII, FIX, FX, and FXI and downregulated hepatic levels of the VK-dependent factors (FII, FVII, FIX, and FX). In addition, C. aronia reduced fecal levels of triglycerides and cholesterol and serum levels triglycerides, cholesterol, LDL-c, and vLDL-c. In conclusion, with the hypocoagulant effect of C. aronia activity involves VK-dependent and non-vitamin K-dependent factors. PRACTICAL APPLICATIONS: In this study, we are reporting for the first time an in vivo hypocoagulant effect of C. aronia in rats. Such effect involved both VK-dependent and independent factors. However, the decrease in the activity and expression of VK-dependent factors was associated with reduced fecal levels of TGs and CHOL and serum levels of TGs, CHOL, LDL-c, and vLDL-c. These data suggest a possible impairment in the VK absorption, transport, or hepatic uptake. These data encourage further pharmacological, translational, and clinical studies to isolate the active ingredients to investigate them at the human level.
Collapse
Affiliation(s)
- Abdullah S Shatoor
- Department of Medicine, Cardiology Section, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Ali Shati
- Department of Biology, College of Science, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Sulaiman M Al Humayed
- Department of Medicine, Cardiology Section, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Abdulaziz K Shatoor
- Department of Medicine, Cardiology Section, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Naif M Alhawiti
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.,King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Sultan A Alqahtani
- King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia.,Department of Basic Medical Sciences, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| |
Collapse
|
126
|
Mineoka Y, Ishii M, Hashimoto Y, Yamashita A, Nakamura N, Fukui M. Platelet to lymphocyte ratio correlates with diabetic foot risk and foot ulcer in patients with type 2 diabetes. Endocr J 2019; 66:905-913. [PMID: 31217392 DOI: 10.1507/endocrj.ej18-0477] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Diabetic foot ulcer is a major complication in patients with diabetes. Platelet-lymphocyte ratio (PLR) has been reported to have a predictive effect to some diabetic complications in recent years. However, it has not been fully elucidated about the relationship between diabetic foot risk or diabetic foot ulcer and PLR in patients with type 2 diabetes. Therefore, we aimed to evaluate this relationship. In this cross-sectional study, we evaluated the relationships between patient's diabetic foot risk with the criteria of the International Working Group on the Diabetic Foot (IWGDF) and prevalent foot ulcer, and PLR in 453 consecutive patients with type 2 diabetes. Propensity score analysis was used to adjust the difference of covariates; age, sex, duration of diabetes, body mass index (BMI), HbA1c, current smoking, hypertension, dyslipidemia, neuropathy, PAD, foot deformity and history of foot ulcers. PLR was higher in patients with high risk diabetic foot or foot ulcer (117 ± 40 vs. 107 ± 31, p = 0.003 and 148 ± 65 vs. 113 ± 56, p < 0.001). A receiver-operating characteristic curve demonstrated that PLR of 130.6 constitutes the cut-off value for prevalent foot ulcer with sensitivity 0.85 and specificity 0.70. Multivariate logistic regression analysis revealed that PLR was positively correlated with prevalent foot ulcer (odds ratio, 1.02; 95% confidence interval 1.01-1.04, p = 0.003) after adjusted for several variables with propensity score analysis. Our results demonstrated that PLR can be a marker for high risk diabetic foot and diabetic foot ulcer in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Yusuke Mineoka
- Department of Internal Medicine, Otsu City Hospital, Otsu, Japan
| | - Michiyo Ishii
- Department of Internal Medicine, Otsu City Hospital, Otsu, Japan
| | - Yoshitaka Hashimoto
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Aki Yamashita
- Department of Nursing, Otsu City Hospital, Otsu, Japan
| | - Naoto Nakamura
- Department of Internal Medicine, Saiseikai Kyoto Hospital, Nagaoka, Japan
| | - Michiaki Fukui
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
127
|
Bakogiannis C, Sachse M, Stamatelopoulos K, Stellos K. Platelet-derived chemokines in inflammation and atherosclerosis. Cytokine 2019; 122:154157. [DOI: 10.1016/j.cyto.2017.09.013] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 07/31/2017] [Accepted: 09/11/2017] [Indexed: 12/16/2022]
|
128
|
Hara T, Phuong PT, Fukuda D, Yamaguchi K, Murata C, Nishimoto S, Yagi S, Kusunose K, Yamada H, Soeki T, Wakatsuki T, Imoto I, Shimabukuro M, Sata M. Protease-Activated Receptor-2 Plays a Critical Role in Vascular Inflammation and Atherosclerosis in Apolipoprotein E-Deficient Mice. Circulation 2019; 138:1706-1719. [PMID: 29700120 DOI: 10.1161/circulationaha.118.033544] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The coagulation system is closely linked with vascular inflammation, although the underlying mechanisms are still obscure. Recent studies show that protease-activated receptor (PAR)-2, a major receptor of activated factor X, is expressed in both vascular cells and leukocytes, suggesting that PAR-2 may contribute to the pathogenesis of inflammatory diseases. Here we investigated the role of PAR-2 in vascular inflammation and atherogenesis. METHODS We generated apolipoprotein E-deficient ( ApoE-/-) mice lacking systemic PAR-2 expression ( PAR-2-/- ApoE-/-). ApoE-/- mice, which lack or express PAR-2 only in bone marrow (BM) cells, were also generated by BM transplantation. Atherosclerotic lesions were investigated after 20 weeks on a Western-type diet by histological analyses, quantitative reverse transcription polymerase chain reaction, and Western blotting. In vitro experiments using BM-derived macrophages were performed to confirm the proinflammatory roles of PAR-2. The association between plasma activated factor X level and the severity of coronary atherosclerosis was also examined in humans who underwent coronary intervention. RESULTS PAR-2-/- ApoE-/- mice showed reduced atherosclerotic lesions in the aortic arch ( P<0.05) along with features of stabilized atherosclerotic plaques, such as less lipid deposition ( P<0.05), collagen loss ( P<0.01), macrophage accumulation ( P<0.05), and inflammatory molecule expression ( P<0.05) compared with ApoE-/- mice. Systemic PAR2 deletion in ApoE-/-mice significantly decreased the expression of inflammatory molecules in the aorta. The results of BM transplantation experiments demonstrated that PAR-2 in hematopoietic cells contributed to atherogenesis in ApoE-/- mice. PAR-2 deletion did not alter metabolic parameters. In vitro experiments demonstrated that activated factor X or a specific peptide agonist of PAR-2 significantly increased the expression of inflammatory molecules and lipid uptake in BM-derived macrophages from wild-type mice compared with those from PAR-2-deficient mice. Activation of nuclear factor-κB signaling was involved in PAR-2-associated vascular inflammation and macrophage activation. In humans who underwent coronary intervention, plasma activated factor X level independently correlated with the severity of coronary atherosclerosis as determined by Gensini score ( P<0.05) and plaque volume ( P<0.01). CONCLUSIONS PAR-2 signaling activates macrophages and promotes vascular inflammation, increasing atherosclerosis in ApoE-/- mice. This signaling pathway may also participate in atherogenesis in humans.
Collapse
Affiliation(s)
- Tomoya Hara
- Departments of Cardiovascular Medicine (T.H., P.T.P., D.F., K.Y., S.N., S.Y., K.K., H.Y., T.S., T.W., M.Sata), Tokushima University Graduate School of Biomedical Sciences, Japan
| | - Pham Tran Phuong
- Departments of Cardiovascular Medicine (T.H., P.T.P., D.F., K.Y., S.N., S.Y., K.K., H.Y., T.S., T.W., M.Sata), Tokushima University Graduate School of Biomedical Sciences, Japan
| | - Daiju Fukuda
- Departments of Cardiovascular Medicine (T.H., P.T.P., D.F., K.Y., S.N., S.Y., K.K., H.Y., T.S., T.W., M.Sata), Tokushima University Graduate School of Biomedical Sciences, Japan.,Cardio-Diabetes Medicine (D.F., M.Shimabukuro), Tokushima University Graduate School of Biomedical Sciences, Japan
| | - Koji Yamaguchi
- Departments of Cardiovascular Medicine (T.H., P.T.P., D.F., K.Y., S.N., S.Y., K.K., H.Y., T.S., T.W., M.Sata), Tokushima University Graduate School of Biomedical Sciences, Japan
| | - Chie Murata
- Human Genetics (C.M., I.I.), Tokushima University Graduate School of Biomedical Sciences, Japan
| | - Sachiko Nishimoto
- Departments of Cardiovascular Medicine (T.H., P.T.P., D.F., K.Y., S.N., S.Y., K.K., H.Y., T.S., T.W., M.Sata), Tokushima University Graduate School of Biomedical Sciences, Japan
| | | | - Kenya Kusunose
- Departments of Cardiovascular Medicine (T.H., P.T.P., D.F., K.Y., S.N., S.Y., K.K., H.Y., T.S., T.W., M.Sata), Tokushima University Graduate School of Biomedical Sciences, Japan
| | - Hirotsugu Yamada
- Departments of Cardiovascular Medicine (T.H., P.T.P., D.F., K.Y., S.N., S.Y., K.K., H.Y., T.S., T.W., M.Sata), Tokushima University Graduate School of Biomedical Sciences, Japan
| | - Takeshi Soeki
- Departments of Cardiovascular Medicine (T.H., P.T.P., D.F., K.Y., S.N., S.Y., K.K., H.Y., T.S., T.W., M.Sata), Tokushima University Graduate School of Biomedical Sciences, Japan
| | - Tetsuzo Wakatsuki
- Departments of Cardiovascular Medicine (T.H., P.T.P., D.F., K.Y., S.N., S.Y., K.K., H.Y., T.S., T.W., M.Sata), Tokushima University Graduate School of Biomedical Sciences, Japan
| | - Issei Imoto
- Human Genetics (C.M., I.I.), Tokushima University Graduate School of Biomedical Sciences, Japan
| | - Michio Shimabukuro
- Cardio-Diabetes Medicine (D.F., M.Shimabukuro), Tokushima University Graduate School of Biomedical Sciences, Japan
| | - Masataka Sata
- Departments of Cardiovascular Medicine (T.H., P.T.P., D.F., K.Y., S.N., S.Y., K.K., H.Y., T.S., T.W., M.Sata), Tokushima University Graduate School of Biomedical Sciences, Japan
| |
Collapse
|
129
|
Raghavan S, Singh NK, Gali S, Mani AM, Rao GN. Protein Kinase Cθ Via Activating Transcription Factor 2-Mediated CD36 Expression and Foam Cell Formation of Ly6C hi Cells Contributes to Atherosclerosis. Circulation 2019; 138:2395-2412. [PMID: 29991487 DOI: 10.1161/circulationaha.118.034083] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Although the role of thrombin in atherothrombosis is well studied, its role in the pathogenesis of diet-induced atherosclerosis is not known. METHODS Using a mouse model of diet-induced atherosclerosis and molecular biological approaches, here we have explored the role of thrombin and its G protein-coupled receptor signaling in diet-induced atherosclerosis. RESULTS In exploring the role of G protein-coupled receptor signaling in atherogenesis, we found that thrombin triggers foam cell formation via inducing CD36 expression, and these events require Par1-mediated Gα12-Pyk2-Gab1-protein kinase C (PKC)θ-dependent ATF2 activation. Genetic deletion of PKCθ in apolipoprotein E (ApoE)-/- mice reduced Western diet-induced plaque formation. Furthermore, thrombin induced Pyk2, Gab1, PKCθ, and ATF2 phosphorylation, CD36 expression, and foam cell formation in peritoneal macrophages of ApoE-/- mice. In contrast, thrombin only stimulated Pyk2 and Gab1 but not ATF2 phosphorylation or its target gene CD36 expression in the peritoneal macrophages of ApoE-/-:PKCθ-/- mice, and it had no effect on foam cell formation. In addition, the aortic root cross-sections of Western diet-fed ApoE-/- mice showed increased Pyk2, Gab1, PKCθ, and ATF2 phosphorylation and CD36 expression as compared with ApoE-/-:PKCθ-/- mice. Furthermore, although the monocytes from peripheral blood and the aorta of Western diet-fed ApoE-/- mice were found to contain more of Ly6Chi cells than Ly6Clo cells, the monocytes from Western diet-fed ApoE-/-:PKCθ-/- mice were found to contain more Ly6Clo cells than Ly6Chi cells. It is interesting to note that the Ly6Chi cells showed higher CD36 expression with enhanced capacity to form foam cells as compared with Ly6Clo cells. CONCLUSIONS These findings reveal for the first time that thrombin-mediated Par1-Gα12 signaling via targeting Pyk2-Gab1-PKCθ-ATF2-dependent CD36 expression might be playing a crucial role in diet-induced atherogenesis.
Collapse
Affiliation(s)
| | - Nikhlesh K Singh
- Department of Physiology, University of Tennessee Health Science Center, Memphis
| | - Sivaiah Gali
- Department of Physiology, University of Tennessee Health Science Center, Memphis
| | - Arul M Mani
- Department of Physiology, University of Tennessee Health Science Center, Memphis
| | - Gadiparthi N Rao
- Department of Physiology, University of Tennessee Health Science Center, Memphis
| |
Collapse
|
130
|
Gutowska K, Formanowicz D, Formanowicz P. Selected Aspects of Tobacco-Induced Prothrombotic State, Inflammation and Oxidative Stress: Modeled and Analyzed Using Petri Nets. Interdiscip Sci 2019; 11:373-386. [PMID: 30584644 PMCID: PMC6660494 DOI: 10.1007/s12539-018-0310-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 11/23/2018] [Accepted: 11/26/2018] [Indexed: 11/27/2022]
Abstract
Many factors, such as endothelial dysfunction, inflammation and hemostatic disturbances, affect formation and progression of atherosclerotic plaque. In our study, we have focused on hemostatic disturbances with particular emphasis on the extrinsic pathways of coagulation. Thrombin is a main enzyme of coagulation and it is engaged in many different subprocesses. It leads to activation of factors of the coagulation cascade, transition of fibrinogen to fibrin monomer, endothelial damage, inflammation, activation of platelets and proliferation. In our study, selected aspects of disorders in prothrombotic states influenced by cigarette smoke have been modeled and analyzed. Tobacco-induced increased tissue factor, which is associated with less plasminogen activator and increased plasminogen activator-1 inhibitor, has been included in the presented model. These disorders together with accompanying inflammatory state are closely related to thrombus formation and cardiovascular disease promotion. The proposed model has been built using Petri nets and the analysis has been based mainly on t-invariants. Using the Petri net theory to model and analyze the investigated phenomena allows to better understand them by revealing some interesting dependencies in the studied biological system. It explains how tobacco smoke affects the analyzed processes and how harmful these effects are.
Collapse
Affiliation(s)
- Kaja Gutowska
- Institute of Computing Science, Poznan University of Technology, Piotrowo 2, 60-965 Poznan, Poland
| | - Dorota Formanowicz
- Department of Clinical Biochemistry and Laboratory Medicine, Poznan University of Medical Sciences, Rokietnicka 8, 60-806 Poznan, Poland
| | - Piotr Formanowicz
- Institute of Computing Science, Poznan University of Technology, Piotrowo 2, 60-965 Poznan, Poland
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| |
Collapse
|
131
|
Pharmacodynamic Effects of Vorapaxar in Patients With and Without Diabetes Mellitus: Results of the OPTIMUS-5 Study. JACC Basic Transl Sci 2019; 4:763-775. [PMID: 31998847 PMCID: PMC6978557 DOI: 10.1016/j.jacbts.2019.07.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 07/31/2019] [Accepted: 07/31/2019] [Indexed: 12/18/2022]
Abstract
Vorapaxar reduces thrombotic cardiovascular events in patients with atherosclerotic disease, with enhanced effects in those with DM. Adjunctive vorapaxar therapy reduces platelet-mediated thrombogenicity without affecting clot kinetics in both patients with and those without DM having prior MI/PAD on dual antiplatelet therapy with aspirin and clopidogrel. The pharmacodynamic effects of vorapaxar occur via selective blockade of the PAR-1 on the platelet membrane without apparent interplay with other platelet signaling pathways. Aspirin withdrawal, which leaves patients on a background of clopidogrel and vorapaxar, increases markers specific to COX-1–mediated blockade, leading to an increase in platelet-mediated global thrombogenicity, particularly among patients with DM.
Vorapaxar reduces thrombotic cardiovascular events at the expense of increased bleeding. However, the differential pharmacodynamic (PD) effects of vorapaxar according to diabetes mellitus (DM) status are unknown. Moreover, although withdrawal of aspirin has emerged as a bleeding reduction strategy, the PD effects of stopping aspirin in patients treated with vorapaxar also are unknown. In this prospective PD investigation, vorapaxar was associated with reduced platelet-mediated thrombogenicity without affecting clot kinetics irrespective of DM status. However, platelet-mediated thrombogenicity increased after aspirin withdrawal, particularly among patients with DM. (Optimizing anti-Platelet Therapy In diabetes MellitUS-5 Study [OPTIMUS-5]; NCT02548650)
Collapse
Key Words
- ADP, adenosine diphosphate
- CAT, collagen-related peptide + adenosine diphosphate + thrombin receptor activating peptide
- CI, confidence interval
- COX, cyclooxygenase
- DAPT, dual antiplatelet therapy
- DM, diabetes mellitus
- LTA, light transmittance aggregometry
- MI, myocardial infarction
- MPA, maximum platelet aggregation
- PAD, peripheral arterial disease
- PAR, protease-activated receptor
- PD, pharmacodynamic
- TRAP, thrombin receptor activating peptide
- TXB2, thromboxane B2
- VASP, vasodilator-stimulated phosphoprotein
- dual antiplatelet therapy
- o.d., once daily
- pharmacodynamics
- platelets
- thrombin
- vorapaxar
Collapse
|
132
|
Baicalin Protects against Thrombin-Induced Cell Injury in Human Umbilical Vein Endothelial Cells. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2187306. [PMID: 31467874 PMCID: PMC6699368 DOI: 10.1155/2019/2187306] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 06/16/2019] [Accepted: 07/11/2019] [Indexed: 01/17/2023]
Abstract
Thrombin plays a pivotal role in the pathogenesis of atherosclerosis. Baicalin, an active flavonoid compound, was shown to attenuate the development of atherosclerosis, but the mechanism remains elusive. In the present study, the role and mechanism of baicalin in thrombin-induced cell injury was investigated in human umbilical vein endothelial cells (HUVECs). Our results showed that baicalin significantly reduced thrombin-induced apoptosis of HUVECs. Additional experiments showed that baicalin inhibited thrombin-induced NF-κB activation and PAR-1 expression. In addition, baicalin decreased thrombin-induced PAR-1 expression by inhibiting ERK pathway. These results indicated that baicalin has protective effects on thrombin-induced cell injury in HUVECs possibly through inhibition of PAR-1 expression and its downstream NF-κB activation, which was mediated by ERK1/2 activation.
Collapse
|
133
|
Rivaroxaban, a specific FXa inhibitor, improved endothelium-dependent relaxation of aortic segments in diabetic mice. Sci Rep 2019; 9:11206. [PMID: 31371788 PMCID: PMC6672013 DOI: 10.1038/s41598-019-47474-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 07/12/2019] [Indexed: 12/27/2022] Open
Abstract
Activated factor X (FXa) plays a central role in the coagulation cascade, while it also mediates vascular function through activation of protease-activated receptors (PARs). Here, we examined whether inhibition of FXa by rivaroxaban, a direct FXa inhibitor, attenuates endothelial dysfunction in streptozotocin (STZ)-induced diabetic mice. Induction of diabetes increased the expression of a major FXa receptor, PAR2, in the aorta (P < 0.05). Administration of rivaroxaban (10 mg/kg/day) to diabetic wild-type (WT) mice for 3 weeks attenuated endothelial dysfunction as determined by acetylcholine-dependent vasodilation compared with the control (P < 0.001), without alteration of blood glucose level. Rivaroxaban promoted eNOSSer1177 phosphorylation in the aorta (P < 0.001). Induction of diabetes to PAR2-deficient (PAR2−/−) mice did not affect endothelial function and eNOSSer1177 phosphorylation in the aorta compared with non-diabetic PAR2−/− mice. FXa or a PAR2 agonist significantly impaired endothelial function in aortic rings obtained from WT mice, but not in those from PAR2−/− mice. FXa promoted JNK phosphorylation (P < 0.01) and reduced eNOSSer1177 phosphorylation (P < 0.05) in human coronary artery endothelial cells (HCAEC). FXa-induced endothelial dysfunction in aortic rings (P < 0.001) and eNOSSer1177 phosphorylation (P < 0.05) in HCAEC were partially ameliorated by a JNK inhibitor. Rivaroxaban ameliorated diabetes-induced endothelial dysfunction. Our results suggest that FXa or PAR2 is a potential therapeutic target.
Collapse
|
134
|
Serologic and Histologic Predictors of Long-Term Renal Outcome in Biopsy-Confirmed IgA Nephropathy (Haas Classification): An Observational Study. J Clin Med 2019; 8:jcm8060848. [PMID: 31207869 PMCID: PMC6617418 DOI: 10.3390/jcm8060848] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/12/2019] [Accepted: 06/12/2019] [Indexed: 02/06/2023] Open
Abstract
Background and objective: The Haas classification of IgA nephropathy should be validated for Asian populations. More detailed and newer predictions regarding renal outcome of IgA nephropathy remains mandatory. Materials: We conducted a retrospective cohort study between January 2003 and December 2013. Clinical, Pathological, and laboratory data were all collected via available medical records. A Mann–Whitney U test was used for continuous variables and the Chi-square test was implemented for categorical variables. A Kaplan–Meier curve was put in place in order to determine patient survival and renal survival. The Youden index and Cox proportional hazard regression were used to investigate the possible factors for renal survival and predictive power. Results: All 272 renal biopsy-confirmed IgAN patients were enrolled for further studies. The univariate analysis showed that risk factors for poor renal outcome included stage 4–5 of Haas classification (HR = 3.67, p < 0.001), a poor baseline renal function (HR = 1.02 and p < 0.001 for higher BUN; HR = 1.14 and p < 0.001 for higher serum creatinine; HR = 0.95, p < 0.001 for higher eGFR), IgG ≤ 907 (HR = 2.29, p = 0.003), C3 ≤ 79.7 (HR = 2.76, p = 0.002), a higher C4 (HR = 1.02, p = 0.026), neutrophil-to-lymphocyte ratio > 2.75 (HR = 2.92, p < 0.001), and a platelet-to-lymphocyte ratio ≥ 16.06 (HR = 2.02, p = 0.012). A routine-checked markers, such as neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio, in order to predict the renal outcome, is recommended. Conclusions: This is the first study to demonstrate that Haas classification is also useful for establishing predictive values in Asian groups. A lower serum IgG (≤907 mg/dL) and serum C3 (≤79.7 mg/dL) were both risk factors for poor renal outcome. Additionally, this is the first study to reveal that serum C4 levels, an NLR > 2.75 and a PLR > 16.06, S could suggest poor renal outcome.
Collapse
|
135
|
Petsophonsakul P, Furmanik M, Forsythe R, Dweck M, Schurink GW, Natour E, Reutelingsperger C, Jacobs M, Mees B, Schurgers L. Role of Vascular Smooth Muscle Cell Phenotypic Switching and Calcification in Aortic Aneurysm Formation. Arterioscler Thromb Vasc Biol 2019; 39:1351-1368. [PMID: 31144989 DOI: 10.1161/atvbaha.119.312787] [Citation(s) in RCA: 217] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Aortic aneurysm is a vascular disease whereby the ECM (extracellular matrix) of a blood vessel degenerates, leading to dilation and eventually vessel wall rupture. Recently, it was shown that calcification of the vessel wall is involved in both the initiation and progression of aneurysms. Changes in aortic wall structure that lead to aneurysm formation and vascular calcification are actively mediated by vascular smooth muscle cells. Vascular smooth muscle cells in a healthy vessel wall are termed contractile as they maintain vascular tone and remain quiescent. However, in pathological conditions they can dedifferentiate into a synthetic phenotype, whereby they secrete extracellular vesicles, proliferate, and migrate to repair injury. This process is called phenotypic switching and is often the first step in vascular pathology. Additionally, healthy vascular smooth muscle cells synthesize VKDPs (vitamin K-dependent proteins), which are involved in inhibition of vascular calcification. The metabolism of these proteins is known to be disrupted in vascular pathologies. In this review, we summarize the current literature on vascular smooth muscle cell phenotypic switching and vascular calcification in relation to aneurysm. Moreover, we address the role of vitamin K and VKDPs that are involved in vascular calcification and aneurysm. Visual Overview- An online visual overview is available for this article.
Collapse
Affiliation(s)
- Ploingarm Petsophonsakul
- From the Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands (P.P., M.F., C.R., L.S.)
| | - Malgorzata Furmanik
- From the Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands (P.P., M.F., C.R., L.S.)
| | - Rachael Forsythe
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (R.F., M.D.)
| | - Marc Dweck
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (R.F., M.D.)
| | - Geert Willem Schurink
- Department of Vascular Surgery (G.W.S., M.J., B.M.), Maastricht University Medical Center (MUMC), Maastricht, the Netherlands
| | - Ehsan Natour
- Department of Cardiovascular Surgery (E.N.), Maastricht University Medical Center (MUMC), Maastricht, the Netherlands.,European Vascular Center Aachen-Maastricht, Maastricht, the Netherlands (E.N., M.J., B.M.)
| | - Chris Reutelingsperger
- From the Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands (P.P., M.F., C.R., L.S.)
| | - Michael Jacobs
- Department of Vascular Surgery (G.W.S., M.J., B.M.), Maastricht University Medical Center (MUMC), Maastricht, the Netherlands.,European Vascular Center Aachen-Maastricht, Maastricht, the Netherlands (E.N., M.J., B.M.)
| | - Barend Mees
- Department of Vascular Surgery (G.W.S., M.J., B.M.), Maastricht University Medical Center (MUMC), Maastricht, the Netherlands.,European Vascular Center Aachen-Maastricht, Maastricht, the Netherlands (E.N., M.J., B.M.)
| | - Leon Schurgers
- From the Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands (P.P., M.F., C.R., L.S.)
| |
Collapse
|
136
|
Wang J, Li X, Pu J, Jin S, Jia L, Li X, Liu F, Yang Y. Mean platelet volume and coronary plaque vulnerability: an optical coherence tomography study in patients with non-ST-elevation acute coronary syndrome. BMC Cardiovasc Disord 2019; 19:128. [PMID: 31142268 PMCID: PMC6542039 DOI: 10.1186/s12872-019-1115-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 05/21/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND The association between mean platelet volume (MPV) and coronary plaque vulnerability in patients with non-ST-elevation ACS (NSTE-ACS) has not been investigated. We performed a retrospective study to evaluate the association between MPV and plaque vulnerability using optical coherence tomography (OCT). METHODS Consecutive NSTE-ACS patients who underwent pre-intervention OCT examination in our center were included in this study. Features of coronary plaques in the culprit arteries were classified as rupture, nonrupture with thin-cap fibroatheroma (TCFA), and nonrupture and non-TCFA. ROC analyses were used to determine the predictive efficacy of MPV for plaque rupture, and multivariate logistic regression analysis was performed to evaluate the potential independent predictors of plaque vulnerability. RESULTS Overall, 94 patients were included in this study. We identified 17 patients with plaque rupture, 10 with nonrupture with TCFA, and 67 with nonrupture and non-TCFA. ROC analyses showed that MPV ≥ 10.5 fL was predictive of plaque rupture in NSTE-ACS patients. Univariate analyses indicated that patients with higher MPV (≥ 10.5 fL) had higher body mass index and poorer lipid profiles compared to those with lower MPV. Moreover, those with higher MPV had higher incidences of plaque rupture and thrombosis (both P < 0.05). Results of multivariate analyses showed that diabetes and higher platelet distribution width (PDW) were independent risk factors of TCFA (P = 0.032 and 0.046, respectively), while diabetes, higher BMI, higher PDW, and higher MPV were independent determinants of plaque rupture in our cohorts (P all < 0.05). CONCLUSIONS Higher MPV is independently associated with higher risk of plaque rupture as evidenced by OCT in our cohort of NSTE-ACS patients.
Collapse
Affiliation(s)
- Jun Wang
- Department of Coronary Heart Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Xing Li
- Department of Coronary Heart Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Jun Pu
- The Renji Hospital of Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Siyu Jin
- Department of Coronary Heart Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Lu Jia
- Department of Coronary Heart Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Xiaomei Li
- Department of Coronary Heart Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Fen Liu
- Department of Coronary Heart Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Yining Yang
- Department of Coronary Heart Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China.
| |
Collapse
|
137
|
Bladder drug mirabegron exacerbates atherosclerosis through activation of brown fat-mediated lipolysis. Proc Natl Acad Sci U S A 2019; 116:10937-10942. [PMID: 31085638 DOI: 10.1073/pnas.1901655116] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Mirabegron (Myrbetriq) is a β3-adrenoreceptor agonist approved for treating overactive bladder syndrome in human patients. This drug can activate brown adipose tissue (BAT) in adult humans and rodents through the β3-adrenoreceptor-mediated sympathetic activation. However, the effect of the mirabegron, approved by the US Food and Drug Administration, on atherosclerosis-related cardiovascular disease is unknown. Here, we show that the clinical dose of mirabegron-induced BAT activation and browning of white adipose tissue (WAT) exacerbate atherosclerotic plaque development. In apolipoprotein E-/- (ApoE -/-) and low-density lipoprotein (LDL) receptor-/- (Ldlr -/-) mice, oral administration of clinically relevant doses of mirabegron markedly accelerates atherosclerotic plaque growth and instability by a mechanism of increasing plasma levels of both LDL-cholesterol and very LDL-cholesterol remnants. Stimulation of atherosclerotic plaque development by mirabegron is dependent on thermogenesis-triggered lipolysis. Genetic deletion of the critical thermogenesis-dependent protein, uncoupling protein 1, completely abrogates the mirabegron-induced atherosclerosis. Together, our findings suggest that mirabegron may trigger cardiovascular and cerebrovascular diseases in patients who suffer from atherosclerosis.
Collapse
|
138
|
Li B, Liu Y, Hu T, Zhang Y, Zhang C, Li T, Wang C, Dong Z, Novakovic VA, Hu T, Shi J. Neutrophil extracellular traps enhance procoagulant activity in patients with oral squamous cell carcinoma. J Cancer Res Clin Oncol 2019; 145:1695-1707. [PMID: 31020419 DOI: 10.1007/s00432-019-02922-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 04/19/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Hypercoagulability is a major cancer-associated complication linked to poor patient prognosis. The production of neutrophil extracellular traps (NETs) is increasingly found to be linked with the development and metastasis of cancer, as well as with thrombi formation in cancer patients. We hypothesized that the neutrophil NET release may be triggered by specific cytokines in oral squamous cell carcinoma (OSCC) patients, thereby predisposing them to a hypercoagulable state. Moreover, we have evaluated the interaction between NETs and endothelial cells (ECs). METHODS NET procoagulant activity was assessed based on fibrin and purified coagulation complex production assays, as well as by measuring coagulation time (CT). We further used confocal microscopy to quantify the exposure of phosphatidylserine (PS), fibrin strands, and cell FVa/Xa binding. RESULTS OSCC patients with stage III/IV exhibited elevated plasma NET levels compared to stage I/II or CTR (all P < 0.05). Neutrophils from OSCC patients are predisposed to amplified NET release compared to those from CTR. Furthermore, depleting IL-8, IL-6, and TNF-α led to a reduction in NET release in the plasma. OSCC NETs increased thrombin and fibrin generation and decreased CT significantly (P < 0.05). When NETs were isolated and used to treat ECs, these cells exhibited disrupted morphology by retracting from their cell-cell junctions and convert to a procoagulant phenotype. These effects could be attenuated by approximately 70% using DNase I. CONCLUSIONS Our findings are consistent with a model wherein OSCC drives a systemic inflammatory state, which, in turn, drives neutrophils to prime and release NETs, which drive the development of a hypercoagulable state. Intervening in this process may be a viable means of disrupting these undesirable coagulation dynamics in stage III/IV OSCC patients.
Collapse
Affiliation(s)
- Baorong Li
- Department of Stomatology, The First Hospital, Harbin Medical University, Harbin, 150001, China
| | - Yingmiao Liu
- Department of Stomatology, The First Hospital, Harbin Medical University, Harbin, 150001, China
| | - Tenglong Hu
- Department of Stomatology, The First Hospital, Harbin Medical University, Harbin, 150001, China. .,Department of Oral Anatomy and Physiology, Stomatology School, Harbin Medical University, Harbin, 150001, China.
| | - Yan Zhang
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, 150001, China
| | - Cong Zhang
- Department of Ultrasound, The First Hospital, Harbin Medical University, Harbin, 150001, China
| | - Tao Li
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, 150001, China
| | - Chunxu Wang
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, 150001, China
| | - Zengxiang Dong
- Department of Cardiology, The First Hospital, Harbin Medical University, Harbin, 150001, China
| | - Valerie A Novakovic
- Department of Research and Surgery, VA Boston Healthcare System, Brigham and Women's Hospital, Harvard Medical School, Boston, 02132, USA
| | - Tianshui Hu
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, 150001, China
| | - Jialan Shi
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, 150001, China. .,Department of Research and Surgery, VA Boston Healthcare System, Brigham and Women's Hospital, Harvard Medical School, Boston, 02132, USA.
| |
Collapse
|
139
|
Papadaki S, Tselepis AD. Nonhemostatic Activities of Factor Xa: Are There Pleiotropic Effects of Anti-FXa Direct Oral Anticoagulants? Angiology 2019; 70:896-907. [PMID: 31010298 DOI: 10.1177/0003319719840861] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Factor Xa (FXa) is the key serine protease of the coagulation cascade as it is the point of convergence of the intrinsic and extrinsic pathways, leading to the formation of thrombin. Factor Xa is an established target of anticoagulation therapy, due to its central role in coagulation. Over the past years, several direct oral anticoagulants (DOACs) targeting FXa have been developed. Rivaroxaban, apixaban, and edoxaban are used in clinical practice for prevention and treatment of thrombotic diseases. Increasing evidence suggests that FXa exerts nonhemostatic cellular effects that are mediated mainly through protease-activated receptors-1 and -2 and are involved in pathophysiological conditions, such as atherosclerosis, inflammation, and fibrosis. Direct inhibition of FXa by DOACs could be beneficial in these conditions. This is a narrative review that focuses on the cellular effects of FXa in various cell types and conditions, as well as on the possible pleiotropic effects of FXa-targeting DOACs.
Collapse
Affiliation(s)
- Styliani Papadaki
- 1 Department of Chemistry, Atherothrombosis Research Centre/Laboratory of Biochemistry, University of Ioannina, Ioannina, Greece
| | - Alexandros D Tselepis
- 1 Department of Chemistry, Atherothrombosis Research Centre/Laboratory of Biochemistry, University of Ioannina, Ioannina, Greece
| |
Collapse
|
140
|
Tsai SF, Wu MJ, Chen CH. Low serum C3 level, high neutrophil-lymphocyte-ratio, and high platelet-lymphocyte-ratio all predicted poor long-term renal survivals in biopsy-confirmed idiopathic membranous nephropathy. Sci Rep 2019; 9:6209. [PMID: 30996263 PMCID: PMC6470169 DOI: 10.1038/s41598-019-42689-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 04/05/2019] [Indexed: 01/05/2023] Open
Abstract
Idiopathic membranous nephropathy (iMN) is the major cause of end-stage renal disease (ESRD). Recent guidelines suggest limiting immunosuppressants only to high risk patients for ESRD. The present study is aimed at identifying new predictors for the renal outcome of iMN patients. We conducted a retrospective cohort study covering a period from January 2003 to December 2013. We enrolled participants who had received their first renal biopsy at our medical center in Taiwan with the diagnosis of iMN. Clinical, pathological and laboratory data were collected from medical records. Analyses with Mann-Whitney U test was used for continuous variables and Chi-square test for categorical variables. The Kaplan-Meier curve was used for the analyses of patient survival and renal survival. Youden index was used for evaluating the performance of a dichotomous diagnostic test for renal and patient outcomes. Cox proportional hazard regression was used to determine factors affecting renal survival.A total of 99 patients with renal biopsy-confirmed idiopathic iMNs were enrolled. C3 level ≤114 mg/dl predicted patient outcome (p < 0.001) with good predictive power (AUC = 0.736). The univariate analysis showed that risk factors for poor renal outcome were older age (HR = 1.04, p = 0.002), high BUN (HR = 1.03, p < 0.001), poor baseline renal function (HR = 1.30 and p < 0.001 for higher serum creatinine; HR = 0.97 and p < 0.001 for higher eGFR; HR = 1.06 and p < 0.001 for urine PCR), C3 ≤ 93.4 mg/dl (HR = 2.15, p = 0.017), NLR > 3.34 (HR = 3.30, p < 0.001) and PLR > 14.48 (HR = 2.54, p = 0.003). Stage of iMN did not fully account for the risk of ESRD. This is the first evidence that serum levels of C3 ≤ 93.4 mg/dl predicted poor renal outcomes with good predictive power. Easily obtained markers, NLR > 3.34 also predicted poor renal outcomes.
Collapse
Affiliation(s)
- Shang-Feng Tsai
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan.,Department of Life Science, Tunghai University, Taichung, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Ming-Ju Wu
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Lilfe Science, National Chung Hsing University, Taichung, Taiwan
| | - Cheng-Hsu Chen
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan. .,Department of Life Science, Tunghai University, Taichung, Taiwan. .,School of Medicine, Chung Shan Medical University, Taichung, Taiwan. .,Department of Lilfe Science, National Chung Hsing University, Taichung, Taiwan.
| |
Collapse
|
141
|
Utility of mean platelet volume to predict the prevalence of coronary artery disease on coronary angiography in patients with stable angina. Coron Artery Dis 2019; 30:615-620. [PMID: 30985481 DOI: 10.1097/mca.0000000000000747] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Approximately 50% of patients with stable angina have coronary artery disease (CAD) on coronary angiography. The mean platelet volume (MPV) has been proposed as a marker that reflects platelet size and reactivity. This study investigated the predictive value of high MPV in patients with stable angina for diagnosing stable CAD. PATIENTS AND METHODS A total of 491 patients with chest pain who underwent selective coronary angiography for suspected CAD were enrolled. The patients were divided into the CAD group and non-CAD group according to angiography. All demographic, laboratory, and angiographic data were collected. RESULTS Patients with MPV in the highest tertile were more likely to have CAD (66.9 vs. 51.0 vs. 35.7% for the highest, middle, and lowest tertiles; P = 0.001), had lower platelet counts (186 ± 48 vs. 199 ± 52 vs. 223 ± 63; P < 0.001), and had higher hemoglobin A1c levels (6.8 ± 1.5 vs. 6.5 ± 1.5 vs. 6.2 ± 1.1; P < 0.001). MPV had a positive correlation with hemoglobin A1c (r = 0.16; P < 0.001). Patients with CAD (n = 248) had higher MPV than those without CAD (n = 243) (11.0 ± 1.0 vs. 10.5 ± 0.9; P < 0.001). MPV was an independent predictor of CAD in patients with stable angina, with an adjusted odds ratio of 1.820 (95% confidence interval: 1.453-2.279; P < 0.001). CONCLUSION The presence of high MPV predicts the prevalence of CAD on coronary angiography in patients with stable angina, and this result may ultimately reduce unnecessary invasive coronary angiography.
Collapse
|
142
|
Bianco M, Gravinese C, Cerrato E, Nuñez-Gil I, Destefanis P, Luciano A, Biscaglia S, Quadri G, Tizzani E, Corleto A, Giolitto S, Lo Savio L, Campo G, Varbella F, Pozzi R. Management of aspirin intolerance in patients undergoing percutaneous coronary intervention. The role of mono-antiplatelet therapy: a retrospective, multicenter, study. Minerva Cardioangiol 2019; 67:94-101. [DOI: 10.23736/s0026-4725.19.04787-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
143
|
Ambrose JA, Bhullar AS. Inflammation and Thrombosis in Coronary Atherosclerosis: Pathophysiologic Mechanisms and Clinical Correlations. EUROPEAN MEDICAL JOURNAL 2019. [DOI: 10.33590/emj/10314648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Inflammation and thrombosis are interrelated processes that are important in the pathogenesis of atherothrombosis. Inflammation is important in both the early and late stages of atherosclerosis, and it involves elements of immune system activation. Low density lipoprotein (LDL) is an important initiator but is not the only one. LDL enters the cell membrane, is modified, and sets into motion a series of events that stimulate the ingress of specific proinflammatory mononuclear cells through the vessel wall. These cells imbibe lipids and form foam cells. Proinflammatory mediators secreted by these cells can eventually lead to intimal thickening and lipid accumulation, forming atherosclerotic plaques. A complex interplay between inflammation, platelet function, and hypercoagulability is a major contributor to the progression from stable to unstable plaque and an acute coronary event. In the later stages of atherosclerosis, inflammatory cells can destabilise certain lipid-rich lesions contributing to symptomatic coronary thrombosis. Thus, thrombosis is the final common pathway for most atherosclerotic complications. Thrombi may also contribute to the asymptomatic rapid progression of atherosclerotic lesions. While antithrombotic agents are important in the treatment of acute coronary syndromes, as well as preventive therapy in high-risk primary prevention and in secondary prevention, the role of specific anti-inflammatory agents is not currently established. If such therapies are to become routine, these anti-inflammatory drugs must significantly reduce events while not adversely affecting a patient’s natural immunity to an extent that erases any potential benefit. This article reviews these two processes with an emphasis on coronary atherosclerosis and its sequelae.
Collapse
Affiliation(s)
- John A. Ambrose
- University of California, San Francisco, San Francisco California, USA
| | | |
Collapse
|
144
|
Tsujino Y, Sakamoto T, Kinoshita K, Nakatani Y, Yamaguchi Y, Kataoka N, Nishida K, Kinugawa K. Edoxaban suppresses the progression of atrial fibrosis and atrial fibrillation in a canine congestive heart failure model. Heart Vessels 2019; 34:1381-1388. [DOI: 10.1007/s00380-019-01377-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 03/08/2019] [Indexed: 10/27/2022]
|
145
|
Posthuma JJ, Posma JJN, van Oerle R, Leenders P, van Gorp RH, Jaminon AMG, Mackman N, Heitmeier S, Schurgers LJ, Ten Cate H, Spronk HMH. Targeting Coagulation Factor Xa Promotes Regression of Advanced Atherosclerosis in Apolipoprotein-E Deficient Mice. Sci Rep 2019; 9:3909. [PMID: 30846818 PMCID: PMC6405752 DOI: 10.1038/s41598-019-40602-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 02/20/2019] [Indexed: 01/09/2023] Open
Abstract
Atherosclerosis is a progressive inflammatory vascular disorder, complicated by plaque rupture and subsequently atherothrombosis. In vitro studies indicate that key clotting proteases, such as factor Xa (FXa), can promote atherosclerosis, presumably mediated through protease activated receptors (PARs). Although experimental studies showed reduced onset of atherosclerosis upon FXa inhibition, the effect on pre-existing plaques has never been studied. Therefore, we investigated effects of FXa inhibition by rivaroxaban on both newly-formed and pre-existing atherosclerotic plaques in apolipoprotein-e deficient (ApoE-/-) mice. Female ApoE-/- mice (age: 8-9 weeks, n = 10/group) received western type diet (WTD) or WTD supplemented with rivaroxaban (1.2 mg/g) for 14 weeks. In a second arm, mice received a WTD for 14 weeks, followed by continuation with either WTD or WTD supplemented with rivaroxaban (1.2 mg/g) for 6 weeks (total 20 weeks). Atherosclerotic burden in aortic arch was assessed by haematoxilin & eosin immunohistochemistry (IHC); plaque vulnerability was examined by IHC against macrophages, collagen, vascular smooth muscle cells (VSMC) and matrix metalloproteinases (MMPs). In addition, PAR1 and -2 expressions and their main activators thrombin and FXa in the plaque were determined in the plaque. Administration of rivaroxaban at human therapeutic concentrations reduced the onset of atherosclerosis (-46%, p < 0.05), and promoted a regression of pre-existing plaques in the carotids (-24%, p < 0.001). In addition, the vulnerability of pre-existing plaques was reduced by FXa inhibition as reflected by reduced macrophages (-39.03%, p < 0.05), enhanced collagen deposition (+38.47%, p < 0.05) and diminished necrotic core (-31.39%, p < 0.05). These findings were accompanied with elevated vascular smooth muscle cells and reduced MMPs. Furthermore, expression of PARs and their activators, thrombin and FXa was diminished after rivaroxaban treatment. Pharmacological inhibition of FXa promotes regression of advanced atherosclerotic plaques and enhances plaque stability. These data suggest that inhibition of FXa may be beneficial in prevention and regression of atherosclerosis, possibly mediated through reduced activation of PARs.
Collapse
Affiliation(s)
- Jelle J Posthuma
- Laboratory for Clinical Thrombosis and Haemostasis, Department of Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Surgery, Amsterdam University Medical Center, Location AMC, Amsterdam, The Netherlands
| | - Jens J N Posma
- Laboratory for Clinical Thrombosis and Haemostasis, Department of Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Rene van Oerle
- Laboratory for Clinical Thrombosis and Haemostasis, Department of Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Peter Leenders
- Department of Pharmacology-Toxicology, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Rick H van Gorp
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Armand M G Jaminon
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Nigel Mackman
- Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Stefan Heitmeier
- Research & Development,Pharmaceuticals, Bayer AG, Wuppertal, Germany
| | - Leon J Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Hugo Ten Cate
- Laboratory for Clinical Thrombosis and Haemostasis, Department of Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Henri M H Spronk
- Laboratory for Clinical Thrombosis and Haemostasis, Department of Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht, The Netherlands.
| |
Collapse
|
146
|
Mokou M, Klein J, Makridakis M, Bitsika V, Bascands JL, Saulnier-Blache JS, Mullen W, Sacherer M, Zoidakis J, Pieske B, Mischak H, Roubelakis MG, Schanstra JP, Vlahou A. Proteomics based identification of KDM5 histone demethylases associated with cardiovascular disease. EBioMedicine 2019; 41:91-104. [PMID: 30826357 PMCID: PMC6443027 DOI: 10.1016/j.ebiom.2019.02.040] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/11/2019] [Accepted: 02/18/2019] [Indexed: 12/14/2022] Open
Abstract
Background The increased prevalence of cardiovascular disease (CVD) indicates a demand for novel therapeutic approaches. Proteome analysis of vascular tissues from animal models and humans with CVD could lead to the identification of novel druggable targets. Methods LC-MS/MS analysis of thoracic aortas from three mouse models of non-diabetic and diabetic (streptozotocin (STZ)-induced) atherosclerosis followed by bioinformatics/pathway analysis was performed. Selected findings were confirmed by proteomics analysis of human vessels from patients with CVD as well as in vitro studies (migration, proliferation, angiogenesis assays) using endothelial (HUVEC) cells. Findings Comparative tissue proteomics of low density lipoprotein receptor deficient (Ldlr−/−) and diabetic Ldlr−/− (Ldlr−/−STZ) with wild type (WT) animals led to the identification of 284 differentially expressed proteins in both models. Among them, 177 proteins were also differentially expressed in diabetic apolipoprotein E deficient (ApoE−/−STZ) mice, suggesting expression changes associated with atherosclerosis independent of the model used. These proteins recapitulated the hallmarks of atherosclerosis. Comparison of these findings with differentially expressed proteins in human vessels with CVD enabled shortlisting of six commonly dysregulated proteins. Among them, lysine-specific demethylase 5D (KDM5D) exhibited pronounced overexpression accompanied by a reduction in the protein levels of its substrate, the trimethylated lysine 4 of histone H3 (H3K4me3), in patients with CVD. Functional interference studies applying a KDM5 inhibitor on HUVEC reduced cell proliferation, migration and tube-forming ability in vitro. Interpretation This high-throughput proteomics strategy identified KDM5 histone demethylases being potentially involved in CVD, possibly by affecting H3K4 methylation. Fund [SysVasc, HEALTH-2013 603288], [ERA-CVD PROACT: ANR-17-ECVD-0006, 01KL1805], [FRM, DEQ20170336759].
Collapse
Affiliation(s)
- Marika Mokou
- Biotechnology Laboratory, Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece; Laboratory of Biology, University of Athens, School of Medicine, Athens, Greece
| | - Julie Klein
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France; Université Toulouse III Paul-Sabatier Toulouse, Toulouse, France
| | - Manousos Makridakis
- Biotechnology Laboratory, Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Vasiliki Bitsika
- Biotechnology Laboratory, Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Jean-Loup Bascands
- INSERM, U1188, Sainte Clotilde, La Réunion, France; Université de La Réunion, Sainte Clotilde, La Réunion, France
| | - Jean Sebastien Saulnier-Blache
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France; Université Toulouse III Paul-Sabatier Toulouse, Toulouse, France
| | - William Mullen
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Michael Sacherer
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Jerome Zoidakis
- Biotechnology Laboratory, Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité University Medicine, Berlin, Germany; German Center for Cardiovascular Research (DZHK), Partner Site, Berlin, Germany; Department of Internal Medicine and Cardiology, German Heart Center, Berlin, Germany; Berlin Institute of Health (BIH), Germany
| | - Harald Mischak
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom; Mosaiques Diagnostics GmbH, Hannover, Germany
| | - Maria G Roubelakis
- Laboratory of Biology, University of Athens, School of Medicine, Athens, Greece
| | - Joost P Schanstra
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France; Université Toulouse III Paul-Sabatier Toulouse, Toulouse, France.
| | - Antonia Vlahou
- Biotechnology Laboratory, Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| |
Collapse
|
147
|
Mahmoud NI, Messiha BAS, Abo-Saif AA, Abdel-Bakky MS. Inhibition of activated factor X; a new pathway in ameliorating carbon tetrachloride-induced liver fibrosis in rats. J Biochem Mol Toxicol 2019; 33:e22287. [PMID: 30719803 DOI: 10.1002/jbt.22287] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 10/13/2018] [Accepted: 12/21/2018] [Indexed: 11/10/2022]
Abstract
Activated factor X has a central role in the coagulation activation and also contributes to chronic inflammation and tissue fibrosis. In this study, rivaroxaban, a direct factor X inhibitor, attenuates liver fibrosis induced by carbon tetrachloride (CCl4 ). Male rats were randomly allocated into three groups: a control group, CCl 4 fibrotic group, and CCl 4 +rivaroxaban (5 mg/kg) group. Liver fibrosis was induced by subcutaneous injection of CCl 4 twice a week for 6 weeks. Rivaroxaban significantly restored the biochemical parameter including inflammatory and fibrosis markers with histopathological evidence using routine and Masson trichrome staining. It reduced also the expression of tissue factor, fibrin, transforming growth factor and α-smooth muscle actin in the liver tissues. This concludes that rivaroxaban attenuates liver injury caused by CCl 4 , at least in part by inhibiting coagulation and proinflammatory activation. In conclusion, rivaroxaban may be used for the management of liver fibrosis.
Collapse
Affiliation(s)
- Nesreen Ishak Mahmoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef, Egypt
| | | | - Ali Ahmed Abo-Saif
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef, Egypt.,Department of Pharmacology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Mohamed Sadek Abdel-Bakky
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef, Egypt.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
148
|
Yuan HQ, Hao YM, Ren Z, Gu HF, Liu FT, Yan BJ, Qu SL, Tang ZH, Liu LS, Chen DX, Jiang ZS. Tissue factor pathway inhibitor in atherosclerosis. Clin Chim Acta 2019; 491:97-102. [PMID: 30695687 DOI: 10.1016/j.cca.2019.01.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 01/24/2019] [Accepted: 01/25/2019] [Indexed: 12/13/2022]
Abstract
Tissue factor pathway inhibitor (TFPI) reduces the development of atherosclerosis by regulating tissue factor (TF) mediated coagulation pathway. In this review, we focus on recent findings on the inhibitory effects of TFPI on endothelial cell activation, vascular smooth muscle cell (VSMC) proliferation and migration, inflammatory cell recruitment and extracellular matrix which are associated with the development of atherosclerosis. Meanwhile, we are also concerned about the impact of TFPI levels and genetic polymorphisms on clinical atherogenesis. This article aims to explain the mechanism in inhibiting the development of atherosclerosis and clinical effects of TFPI, and provide new ideas for the clinical researches and mechanism studies of atherothrombosis.
Collapse
Affiliation(s)
- Hou-Qin Yuan
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Ya-Meng Hao
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Zhong Ren
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Hong-Feng Gu
- Department of Physiology, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Feng-Tao Liu
- Center of Functional Laboratory, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 42100, PR China
| | - Bin-Jie Yan
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Shun-Lin Qu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Zhi-Han Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Lu-Shan Liu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China
| | - Da-Xing Chen
- Division of Transplantation Immunology and Mucosal Biology, Faculty of Life Sciences and Medicine, King's College London, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Zhi-Sheng Jiang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, PR China.
| |
Collapse
|
149
|
Mohamed AAB, Elnady HM, Alhewaig HK, Moslem Hefny H, Khodery A. The mean platelet volume and plateletcrit as predictors of short-term outcome of acute ischemic stroke. THE EGYPTIAN JOURNAL OF NEUROLOGY, PSYCHIATRY AND NEUROSURGERY 2019; 55:4. [PMID: 30686914 PMCID: PMC6325094 DOI: 10.1186/s41983-018-0035-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 10/23/2018] [Indexed: 02/07/2023] Open
Abstract
Background Activation of the platelet plays an important role in the process of atherosclerosis. Mean platelet volume (MPV) is significantly associated with the poor outcome of acute ischemic stroke while the results of studies about the relationship between plateletcrit (PCT) and stroke outcome were inconsistent. The aim of this work is to determine whether an association exists between MPV and plateletcrit (PCT) and outcome of acute ischemic stroke. Methods We examined 157 patients with ischemic stroke, admitted to the Sohag University Hospital. The diagnosis of stroke was performed clinically according to The World Health Organization and confirmed by brain CT and MRI when needed. Platelet indices including MPV and PCT were assessed immediately (within 2 h) after admission. After 3 months, the functional outcome was assessed using the modified Rankin Scale (mRS) with assessment of the relationship between platelet indices and stroke outcome. Results About 50% of the participants have favorable outcome. MPV was significantly higher in the unfavorable group (10.4 ± 2.3 fL) than in the favorable one (8.7 ± 1.3 fL) (P < 0. 001). MPV was an independent predictor of poor short-term outcome of acute stroke after controlling for confounders like diabetes mellitus. The mean PCT was significantly higher in the unfavorable group (0.28 ± 0.1%) than in the favorable one (0.25 ± 0.1%) (P = 0. 04) but not considered as an independent predictor of poor short-term outcome of acute stroke. Conclusions MPV and PCT were significantly correlated with poor functional outcome, only MPV was an independent predictor of poor short-term outcome of acute stroke after controlling for confounders like DM, and these platelet indices can be used as a prognostic tool.
Collapse
Affiliation(s)
- Al-Amir Bassiouny Mohamed
- 1Department of Neurology and Psychological Medicine, Faculty of Medicine, Sohag University Hospital, Sohag, Egypt
| | - Hassan Mohamed Elnady
- 1Department of Neurology and Psychological Medicine, Faculty of Medicine, Sohag University Hospital, Sohag, Egypt
| | - Hazem Kamal Alhewaig
- 1Department of Neurology and Psychological Medicine, Faculty of Medicine, Sohag University Hospital, Sohag, Egypt
| | - Hesham Moslem Hefny
- 2Department of Clinical Pathology, Faculty of Medicine, Sohag University Hospital, Sohag, Egypt
| | - Ashraf Khodery
- 2Department of Clinical Pathology, Faculty of Medicine, Sohag University Hospital, Sohag, Egypt
| |
Collapse
|
150
|
Eggebrecht L, Prochaska JH, Tröbs SO, Schwuchow-Thonke S, Göbel S, Diestelmeier S, Schulz A, Arnold N, Panova-Noeva M, Koeck T, Rapp S, Gori T, Lackner KJ, Ten Cate H, Münzel T, Wild PS. Direct oral anticoagulants and vitamin K antagonists are linked to differential profiles of cardiac function and lipid metabolism. Clin Res Cardiol 2019; 108:787-796. [PMID: 30604046 DOI: 10.1007/s00392-018-1408-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/17/2018] [Indexed: 01/12/2023]
Abstract
BACKGROUND Experimental data indicate that direct acting oral anticoagulants (DOAC) and vitamin K antagonists (VKA) may exert differential effects on cardiovascular disease. METHODS Data from the prospective, observational, single-center MyoVasc Study were used to examine associations of DOAC as compared to VKA with subclinical markers of cardiovascular disease, cardiac function, and humoral biomarkers in heart failure (HF). RESULTS Multivariable analysis adjusted for age, sex, traditional cardiovascular risk factors, comorbidities, and medications with correction for multiple testing demonstrated that DOAC therapy was among all investigated parameters an independent significant predictor of better diastolic function (E/E': β - 0.24 [- 0.36/- 0.12]; P < 0.0001) and higher levels of ApoA1 (β + 0.11 g/L [0.036/0.18]; P = 0.0038) compared to VKA therapy. In propensity score-weighted analyses, the most pronounced differences between DOAC and VKA-based therapy were also observed for E/E' (∆ - 2.36) and ApoA1 (∆ + 0.06 g/L). Sensitivity analyses in more homogeneous subsamples of (i) individuals with AF and (ii) individuals with asymptomatic HF confirmed the consistency and robustness of these findings. In the comparison of factor IIa and Xa-directed oral anticoagulation, no differences were observed regarding cardiac function (E/E' ratio: βIIa inhibitor - 0.22 [- 0.36/- 0.08] vs. βXa inhibitor - 0.24 [- 0.37/- 0.11]) and lipid metabolism (ApoA1: βIIa inhibitor 0.10 [0.01/0.18] vs. βXa inhibitor 0.12 [0.04/0.20]) compared to VKA therapy. CONCLUSION This study provides the first evidence for differential, non-conventional associations of oral anticoagulants on cardiac function and lipid metabolism in humans. The potentially beneficial effect of DOACs in the highly vulnerable population of HF individuals needs to be further elucidated and may have implications for individually tailored anticoagulation therapy.
Collapse
Affiliation(s)
- Lisa Eggebrecht
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.,Center for Translational Vascular Biology (CTVB), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Jürgen H Prochaska
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.,Center for Translational Vascular Biology (CTVB), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Sven-Oliver Tröbs
- Center for Translational Vascular Biology (CTVB), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, Mainz, Germany.,Center for Cardiology-Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Sören Schwuchow-Thonke
- Center for Translational Vascular Biology (CTVB), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, Mainz, Germany.,Center for Cardiology-Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Sebastian Göbel
- Center for Translational Vascular Biology (CTVB), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, Mainz, Germany.,Center for Cardiology-Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Simon Diestelmeier
- Center for Translational Vascular Biology (CTVB), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, Mainz, Germany.,Center for Cardiology-Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Andreas Schulz
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.,Center for Translational Vascular Biology (CTVB), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Natalie Arnold
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.,Center for Translational Vascular Biology (CTVB), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Marina Panova-Noeva
- Center for Translational Vascular Biology (CTVB), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Thomas Koeck
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, Mainz, Germany
| | - Steffen Rapp
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, Mainz, Germany
| | - Tommaso Gori
- Center for Translational Vascular Biology (CTVB), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, Mainz, Germany.,Center for Cardiology-Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Karl J Lackner
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, Mainz, Germany.,Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Mainz, Johannes Gutenberg-University, Mainz, Germany
| | - Hugo Ten Cate
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Thrombosis Expertise Center Maastricht, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, 6200, Maastricht, The Netherlands
| | - Thomas Münzel
- Center for Translational Vascular Biology (CTVB), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Center for Cardiology-Cardiology I, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Philipp Sebastian Wild
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany. .,Center for Translational Vascular Biology (CTVB), University Medical Center Mainz, Johannes Gutenberg-University Mainz, Mainz, Germany. .,German Center for Cardiovascular Research (DZHK), Partner Site Rhine Main, Mainz, Germany. .,Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
| |
Collapse
|