1
|
Brook R, Suleiman M, Rigano J, Lui B, Nandurkar H, Ho P, Lim HY. Tissue factor pathway inhibitor levels and atherothrombotic events in patients with chronic kidney disease or diabetes. J Thromb Thrombolysis 2025; 58:391-400. [PMID: 40064839 PMCID: PMC12009229 DOI: 10.1007/s11239-025-03077-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/07/2025] [Indexed: 04/20/2025]
Abstract
Increased tissue factor pathway inhibitor (TFPI) has been associated with cardiovascular disease (CVD). We aim to evaluate the predictive capability of TFPI for atherothrombotic events (ATE) in patients with chronic kidney disease (CKD) and diabetes. A prospective observational study was performed at Northern Health, Australia. Patients with CKD (estimated glomerular filtration ratio (eGFR) < 30 ml/min/1.73m2) and/or diabetes were recruited. Baseline total TFPI was measured and the median follow-up was 3.35 years. All patients with egfr < 30 ml/min/1.73m2 were analysed as CKD cohort while the diabetes cohort analysis excluded those with egfr < 30 ml/min/1.73m2. The primary outcome was ATE (myocardial infarction, stroke/transient ischaemic attack, critical limb ischaemia or sudden cardiac death). 220 patients were recruited, median age 63.5 years (IQR 51.0, 72.5) and 59.1% males (n = 130). No differences were seen in TFPI levels between the CKD (n = 77) and diabetes (n = 143) cohorts (35.4 vs. 36.4 ng/mL, p = 0.44). TFPI did not correlate with creatinine or HbA1c levels. 46 episodes of ATE were captured (6.69/100-person years (100PY)), with a higher rate in the CKD compared to the diabetes cohort (16.03/100PY vs. 2.53/100PY). In the CKD cohort, those who experienced ATE had higher TFPI with an optimal calculated cut-off (61.36ng/mL) associated with a subhazard ratio of 3.23 (95%CI 1.59-6.57). In the diabetes cohort however, TFPI was not significantly higher in those who experience ATE (40.1 vs. 34.4ng/mL, p = 0.35). We found elevated TFPI may predict prospective ATE, particularly in patients with CKD. While further validation studies are required, these findings highlight that coagulation changes may differ between high-risk CVD populations.
Collapse
Affiliation(s)
- Rowena Brook
- Northern Clinical Diagnostics and Thrombovascular Research (NECTAR), Northern Health, Melbourne, Australia.
- Department of Haematology, Northern Health, Melbourne, Australia.
- Northern Pathology Victoria, Melbourne, Australia.
- Department of Medicine- Northern Health, University of Melbourne, Melbourne, Victoria, Australia.
| | - Mani Suleiman
- Research Development and Governance Unit, Northern Health, Melbourne, VIC, Australia
| | - Joseph Rigano
- Northern Clinical Diagnostics and Thrombovascular Research (NECTAR), Northern Health, Melbourne, Australia
- Northern Pathology Victoria, Melbourne, Australia
| | - Brandon Lui
- Northern Clinical Diagnostics and Thrombovascular Research (NECTAR), Northern Health, Melbourne, Australia
- Department of Haematology, Northern Health, Melbourne, Australia
| | - Harshal Nandurkar
- Department of Haematology, Northern Health, Melbourne, Australia
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Prahlad Ho
- Northern Clinical Diagnostics and Thrombovascular Research (NECTAR), Northern Health, Melbourne, Australia
- Department of Haematology, Northern Health, Melbourne, Australia
- Northern Pathology Victoria, Melbourne, Australia
- Department of Medicine- Northern Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Hui Yin Lim
- Northern Clinical Diagnostics and Thrombovascular Research (NECTAR), Northern Health, Melbourne, Australia
- Department of Haematology, Northern Health, Melbourne, Australia
- Northern Pathology Victoria, Melbourne, Australia
- Department of Medicine- Northern Health, University of Melbourne, Melbourne, Victoria, Australia
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
2
|
Benkhoff M, Alde K, Ehreiser V, Dahlmanns J, Metzen D, Haurand JM, Duse DA, Jung C, Kelm M, Petzold T, Polzin A. Thromboinflammation is associated with clinical outcome after ST-elevation myocardial infarction. Blood Adv 2024; 8:5581-5589. [PMID: 39226457 PMCID: PMC11541696 DOI: 10.1182/bloodadvances.2024014273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/05/2024] Open
Abstract
ABSTRACT Platelets are crucial in thrombus formation during ST-elevation myocardial infarction (STEMI). In addition, they also play an important role in postischemic thromboinflammation, which is determined by the interplay between activated platelets and neutrophils. The latter form neutrophil extracellular traps, which are detectable in plasma as citrullinated histone H3-deoxyribonucleic acid-DNA complexes. Prediction of the risk of recurrent events is important in precision medicine. Therefore, we investigated whether circulating thromboinflammatory markers predict clinical outcome after STEMI. We performed a prospective, multicentric, observational, all-comer study of patients with STEMI (n = 361). Thromboinflammation, measured as H3Cit-DNA complexes, was assessed on day 1 after presentation with STEMI as well as 5 days and 6 months after STEMI by enzyme-linked immunosorbent assay. Twelve months of clinical follow-up was conducted. Multivariate analysis was performed investigating which variables were independently associated with major adverse cardiac events (MACEs). Patients were aged 64 ± 12 years; 80% were male; and 40% had diabetes mellitus. Thromboinflammation was enhanced during index hospitalization compared with 6-months follow-up (137.4 ± 100.0 μg/L vs 53.7 ± 54.7 μg/L; P < .001). Additionally, patients within the highest tertile of thromboinflammation at day 1 after STEMI showed worse outcome during follow-up (hazard ratio, 2.57; 95% confidence interval, 1.72-3.85; P < .001). Receiver operating characteristic analysis revealed a cutoff value of 219.3 μg/L. In multivariate logistic regression analysis, thromboinflammation was independently associated with outcome after STEMI. To sum it up, thromboinflammation is enhanced in STEMI. It identifies patients at high risk of MACE. Therefore, thromboinflammation might be a promising target and marker in precision medicine. The trial was registered at www.clinicaltrials.gov as #NCT03539133.
Collapse
Affiliation(s)
- Marcel Benkhoff
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Analytical Chemistry, University of Vienna, Vienna, Austria
| | - Karin Alde
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Vincent Ehreiser
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité University Hospital Berlin, Campus Benjamin Franklin, Berlin, Germany
- German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany
- Friede Springer-Centre of Cardiovascular Prevention at Charité, Charité-University Medicine Berlin, Berlin, Germany
| | - Jana Dahlmanns
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Daniel Metzen
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jean M. Haurand
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Dragos Andrei Duse
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Christian Jung
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf, Düsseldorf, Germany
| | - Malte Kelm
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf, Düsseldorf, Germany
| | - Tobias Petzold
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité University Hospital Berlin, Campus Benjamin Franklin, Berlin, Germany
- German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany
- Friede Springer-Centre of Cardiovascular Prevention at Charité, Charité-University Medicine Berlin, Berlin, Germany
| | - Amin Polzin
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf, Düsseldorf, Germany
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
3
|
Gastoł J, Paszek E, Bryk-Wiązania A, Matejko B, Undas A. Good metabolic control is associated with decreased circulating factor VIIa- antithrombin complexes in type 2 diabetes: a cross-sectional study. Cardiovasc Diabetol 2024; 23:398. [PMID: 39501309 PMCID: PMC11536800 DOI: 10.1186/s12933-024-02480-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/21/2024] [Indexed: 11/09/2024] Open
Abstract
BACKGROUND Diabetes is associated with a prothrombotic state that contributes to cardiovascular (CV) events in type 2 diabetes (T2DM). Activated factor VII (FVIIa)- antithrombin (AT) complexes are indicative of tissue factor (TF) exposure and have been associated with thromboembolic risk in coronary artery disease. To our knowledge there have been no reports on FVIIa-AT complexes in T2DM, therefore we assessed factors that determine FVIIa-AT complexes in this disease and the impact of higher complexes on a prothrombotic state. METHODS In 108 T2DM patients (mean age 63.8 years, 52.8% men, median HbA1c of 6.9 [interquartile range 6.1-8.2] %) and 83 age- and sex-matched non-diabetic subjects, we measured FVIIa-AT complexes. Metabolic control of T2DM involved fasting glucose, glycated hemoglobin (HbA1c), albumin/creatinine ratio (ACR), and lipid levels. To characterize a prothrombotic state, we determined thrombin generation parameters, fibrinolysis markers, and plasma fibrin clot properties. RESULTS FVII-AT complexes in T2DM patients were similar to controls (73.6 [59.4-91.7] vs. 79.6 [59.2-97.1]pM, respectively, p = 0.30). The T2DM patients with FVIIa-AT in the top vs. the bottom quartile had a larger prevalence of active smoking and insulin use, along with higher fasting glucose (+ 36.4%), HbA1c (+ 27.4%), ACR (+ 72.8%), total cholesterol (+ 34.5%), and LDL-cholesterol (+ 80%). FVIIa-AT complexes showed no associations with in vitro thrombin generation potential, plasma fibrin clot properties, or fibrinolysis variables. On multivariable analysis HbA1c, ACR, and total cholesterol remained independently associated with FVIIa-AT complexes in T2DM. CONCLUSIONS This is the first study to show that in T2DM higher FVIIa-AT complexes are associated with markers of dyslipidemia and glycemia control, indicating that TF-induced coagulation activation could be suppressed by achieving treatment targets.
Collapse
Affiliation(s)
- Joanna Gastoł
- Metabolic Diseases and Diabetology Clinical Department, University Hospital, Kraków, Poland
- Department of Metabolic Diseases, Jagiellonian University Medical College, Kraków, Poland
| | - Elżbieta Paszek
- Clinical Department of Interventional Cardiology, John Paul II Hospital, Kraków, Poland
- Department of Thromboembolic Disorders, Institute of Cardiology, Jagiellonian University Medical College, 80 Pradnicka St, 31-202, Kraków, Poland
| | - Agata Bryk-Wiązania
- Department of Endocrinology, Jagiellonian University Medical College, Kraków, Poland
- Department of Endocrinology, Oncological Endocrinology and Nuclear Medicine, University Hospital, Kraków, Poland
| | - Bartłomiej Matejko
- Metabolic Diseases and Diabetology Clinical Department, University Hospital, Kraków, Poland
- Department of Metabolic Diseases, Jagiellonian University Medical College, Kraków, Poland
| | - Anetta Undas
- Department of Thromboembolic Disorders, Institute of Cardiology, Jagiellonian University Medical College, 80 Pradnicka St, 31-202, Kraków, Poland.
- Cracow Center for Medical Research and Technology, John Paul II Hospital, Kraków, Poland.
| |
Collapse
|
4
|
Heath SP, Hermanns VC, Coucha M, Abdelsaid M. SARS-CoV-2 Spike Protein Exacerbates Thromboembolic Cerebrovascular Complications in Humanized ACE2 Mouse Model. Transl Stroke Res 2024:10.1007/s12975-024-01301-5. [PMID: 39354270 PMCID: PMC11973837 DOI: 10.1007/s12975-024-01301-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/09/2024] [Accepted: 09/26/2024] [Indexed: 10/03/2024]
Abstract
COVID-19 increases the risk for acute ischemic stroke, yet the molecular mechanisms are unclear and remain unresolved medical challenges. We hypothesize that the SARS-CoV-2 spike protein exacerbates stroke and cerebrovascular complications by increasing coagulation and decreasing fibrinolysis by disrupting the renin-angiotensin-aldosterone system (RAAS). A thromboembolic model was induced in humanized ACE2 knock-in mice after one week of SARS-CoV-2 spike protein injection. hACE2 mice were treated with Losartan, an angiotensin receptor (AT1R) blocker, immediately after spike protein injection. Cerebral blood flow and infarct size were compared between groups. Vascular-contributes to cognitive impairments and dementia was assessed using a Novel object recognition test. Tissue factor-III and plasminogen activator inhibitor-1 were measured using immunoblotting to assess coagulation and fibrinolysis. Human brain microvascular endothelial cells (HBMEC) were exposed to hypoxia with/without SARS-CoV-2 spike protein to mimic ischemic conditions and assessed for inflammation, RAAS balance, coagulation, and fibrinolysis. Our results showed that the SARS-CoV-2 spike protein caused an imbalance in the RAAS that increased the inflammatory signal and decreased the RAAS protective arm. SARS-CoV-2 spike protein increased coagulation and decreased fibrinolysis when coincident with ischemic insult, which was accompanied by a decrease in cerebral blood flow, an increase in neuronal death, and a decline in cognitive function. Losartan treatment restored RAAS balance and reduced spike protein-induced effects. SARS-CoV-2 spike protein exacerbates inflammation and hypercoagulation, leading to increased neurovascular damage and cognitive dysfunction. However, the AT1R blocker, Losartan, restored the RAAS balance and reduced COVID-19-induced thromboembolic cerebrovascular complications.
Collapse
Affiliation(s)
- Stan P Heath
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, GA, USA
| | - Veronica C Hermanns
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, GA, USA
| | - Maha Coucha
- Department of Pharmaceutical Sciences, School of Pharmacy, South University, Savannah, GA, USA
| | - Mohammed Abdelsaid
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, GA, USA.
- Biomedical Sciences Department, Mercer University School of Medicine, 1250 E 66th Street | Savannah, 31404, Macon, GA, United States.
| |
Collapse
|
5
|
Marwaha S, Papadakis M. Anabolic steroids in athletes: the interplay of hormones and inflammation leading to the heart's vulnerability. Eur J Prev Cardiol 2024; 31:1477-1479. [PMID: 39096270 DOI: 10.1093/eurjpc/zwae253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 08/05/2024]
Affiliation(s)
- Sarandeep Marwaha
- City St. George's, University of London, St. George's University Hospitals NHS Foundation Trust, UK
| | - Michael Papadakis
- City St. George's, University of London, St. George's University Hospitals NHS Foundation Trust, UK
| |
Collapse
|
6
|
Puspitasari YM, Ministrini S, Han J, Karch C, Prisco F, Liberale L, Bengs S, Akhmedov A, Montecucco F, Beer JH, Lüscher TF, Bongiovanni D, Camici GG. Hutchinson-Gilford progeria syndrome mice display accelerated arterial thrombus formation and increased platelet reactivity. Thromb Res 2024; 241:109100. [PMID: 39032390 DOI: 10.1016/j.thromres.2024.109100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
INTRODUCTION Hutchinson-Gilford Progeria Syndrome (HGPS) is an ultra-rare premature aging genetic disorder caused by a point mutation in the lamin A gene, LMNA. Children with HGPS display short lifespans and typically die due to myocardial infarction or ischemic stroke, both acute cardiovascular events that are tightly linked to arterial thrombosis. Despite this fact, the effect of the classic HGPS LMNA gene mutation on arterial thrombosis remains unknown. METHODS Heterozygous LmnaG609G knock-in (LmnaG609G/+) mice, yielding an equivalent classic mutation observed in HGPS patients (c.1824C>T; pG608G mutation in the human LMNA gene) and corresponding wild-type (WT) control littermates underwent photochemically laser-induced carotid injury to trigger thrombosis. Coagulation and fibrinolytic factors were measured. Furthermore, platelet activation and reactivity were investigated. RESULTS LmnaG609G/+ mice displayed accelerated arterial thrombus formation, as underlined by shortened time to occlusion compared to WT littermates. Levels of factors involved in the coagulation and fibrinolytic system were comparable between groups, while LmnaG609G/+ animals showed higher plasma levels of thrombin-antithrombin complex and lower levels of antithrombin. Bone marrow analysis showed larger megakaryocytes in progeric mice. Lastly, enhanced platelet activation upon adenosine diphosphate, collagen-related peptide, and thrombin stimulation was observed in LmnaG609G/+ animals compared to the WT group, indicating a higher platelet reactivity in progeric animals. CONCLUSIONS LMNA mutation in HGPS mice accelerates arterial thrombus formation, which is mediated, at least in part, by enhanced platelet reactivity, which consequently augments thrombin generation. Given the wide spectrum of antiplatelet agents available clinically, further investigation is warranted to consider the most suitable antiplatelet regimen for children with HGPS to mitigate disease mortality and morbidity.
Collapse
Affiliation(s)
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Internal Medicine, Angiology and Atherosclerosis, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Jiaying Han
- Department of Internal Medicine I, School of Medicine, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - Caroline Karch
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Francesco Prisco
- Laboratory for Animal Model Pathology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Switzerland
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Susan Bengs
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Alexander Akhmedov
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Jürg H Beer
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Department of Internal Medicine, Cantonal Hospital of Baden, Baden, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Department of Cardiology, Royal Brompton & Harefield Hospitals, National Heart & Lung Institute, Imperial College, London, United Kingdom
| | - Dario Bongiovanni
- Department of Internal Medicine I, Cardiology, University Hospital Augsburg, University of Augsburg, Augsburg, Germany; Department of Cardiovascular Medicine, Humanitas Clinical and Research Center IRCCS and Humanitas University, Rozzano, Milan, Italy
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Department of Research and Education, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
7
|
Moruzzi S, Castagna A, Spizzo M, Udali S, Pattini P, Pizzolo F, Friso S, Martinelli N. Activated Factor VII-Antithrombin Complex, a Biomarker of Tissue Factor-Related Pathways in Different Clinical Settings: A Narrative Review from Cardiovascular Diseases to Cancer. Diagnostics (Basel) 2024; 14:1711. [PMID: 39202199 PMCID: PMC11354109 DOI: 10.3390/diagnostics14161711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/02/2024] [Accepted: 08/03/2024] [Indexed: 09/03/2024] Open
Abstract
Tissue factor (TF) is a transmembrane glycoprotein that represents the fundamental physiological initiator of the coagulation cascade through its interaction with factor VII. TF belongs to the cytokine receptor protein superfamily and contributes to the transduction of cellular signaling. Therefore, TF-related pathways are involved in multiple pathophysiological processes, not only in coagulation/thrombosis but in a wider mechanisms' panorama, ranging from infective to neoplastic diseases. Consistently, the measurement of TF activity could have a diagnostic and/or prognostic meaning in different clinical conditions. However, the transmembrane localization, the expression on different cellular types and circulating extracellular vesicles, and the different conformations (encrypted and decrypted) and variants (such as the soluble alternatively spliced TF) hamper TF assessment in clinical practice. The activated factor VII-antithrombin (FVIIa-AT) complex is proposed as an indirect biomarker of the TF-FVIIa interaction and, consequently, of the functionally active TF expression. In this narrative review, we evaluate the clinical studies investigating the role of plasma concentration of FVIIa-AT in health and disease. Although without conclusive data, high FVIIa-AT concentrations predict the worst clinical outcomes in different pathologic conditions, such as cardiovascular disease and cancer, thereby suggesting that overactivation of TF-related pathways may play an unfavorable role in various clinical settings.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Nicola Martinelli
- Department of Medicine, University of Verona, 37134 Verona, Italy; (S.M.); (A.C.); (M.S.); (S.U.); (P.P.); (F.P.); (S.F.)
| |
Collapse
|
8
|
Peng C, Li J, Chen Y, Zhang HR, Li TX, Jiang YH, Yang XY, Zhao Y. PCSK9 aggravated carotid artery stenosis in ApoE -/- mice by promoting the expression of tissue factors in endothelial cells via the TLR4/NF-κB pathway. Biochem Pharmacol 2024; 225:116314. [PMID: 38797271 DOI: 10.1016/j.bcp.2024.116314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 05/09/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
Atherosclerosis, a chronic inflammatory disease, is the most relevant cause of carotid artery stenosis. Vascular endothelial cells (ECs) play a significant role in the development of atherosclerosis. In this chronic inflammatory environment, we aimed to investigate whether PCSK9 could mitigate atherosclerosis progression by reducing tissue factor expression in ECs via in vivo and in vitro assays. In vivo, we investigated the effect of PCSK9 inhibition on preventing atherosclerotic lesion formation in ApoE-/- mice fed a western diet. The results showed that inhibiting PCSK9 could significantly downregulate the protein expression of tissue factor (TF) in ECs to reduce the area of atherosclerotic plaques. In vitro, we incubated human umbilical vein endothelial cells (HUVECs) with lipopolysaccharide (LPS). We found that LPS-induced TF elevation was suppressed by a PCSK9 inhibitor at both the mRNA and protein levels and that the TLR4/NF-κB pathway was also suppressed by a PCSK9 inhibitor. With respect to plasma samples from patients with carotid artery stenosis, we also demonstrated that the expression of TF was positively correlated with that of PCSK9. Thus, in addition to regulating lipid metabolism, the regulation of endothelial cell TF expression through the TLR4/NF-κB pathway may be a potential mechanism of PCSK9 in promoting atherosclerotic carotid stenosis.
Collapse
Affiliation(s)
- Chao Peng
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Jian Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Yan Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Heng-Rui Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Tian-Xing Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Yu-Hang Jiang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Xin-Yu Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Yan Zhao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, PR China.
| |
Collapse
|
9
|
Dregoesc MI, Tercan H, Țigu AB, Bekkering S, Joosten LAB, Netea MG, van Deuren RC, Hoischen A, Riksen NP, Iancu AC. Clonal hematopoiesis is associated with cardiovascular events in patients with stable coronary artery disease. iScience 2024; 27:109472. [PMID: 38558938 PMCID: PMC10981089 DOI: 10.1016/j.isci.2024.109472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 01/23/2024] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Abstract
Clonal hematopoiesis (CH) is a risk factor for atherosclerotic cardiovascular disease, but the impact of smaller clones and the effect on inflammatory parameters is largely unknown. Using ultrasensitive single-molecule molecular inversion probe sequencing, we evaluated the association between CH and a first major adverse cardiovascular event (MACE) in patients with angiographically documented stable coronary artery disease (CAD) and no history of acute ischemic events. CH was associated with an increased rate of MACE at four years follow-up. The size of the clone predicted MACE at an optimal cut-off value of 1.07% variant allele frequency (VAF). Mutation carriers had no change in monocytes subsets or cytokine production capacity but had higher levels of circulating tissue factor, matrilysin, and proteinase-activated receptor-1. Our study identified CH driver mutations with a VAF as small as 1.07% as a residual cardiovascular risk factor and identified potential biomarkers and therapeutic targets for patients with stable CAD.
Collapse
Affiliation(s)
- Mihaela I. Dregoesc
- “Iuliu Hatieganu” University of Medicine and Pharmacy, Department of Cardiology –“Niculae Stăncioiu” Heart Institute, 19-21 Calea Moților, 400001 Cluj-Napoca, Romania
| | - Helin Tercan
- Radboud University Medical Center, Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA Nijmegen, the Netherlands
| | - Adrian B. Țigu
- MEDFUTURE Research Center for Advanced Medicine, Department of Translational Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 4-6 Louis Pasteur, 400349 Cluj-Napoca, Romania
| | - Siroon Bekkering
- Radboud University Medical Center, Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA Nijmegen, the Netherlands
| | - Leo AB. Joosten
- Radboud University Medical Center, Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA Nijmegen, the Netherlands
- Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, 4-6 Louis Pasteur, 400349 Cluj-Napoca, Romania
| | - Mihai G. Netea
- Radboud University Medical Center, Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA Nijmegen, the Netherlands
- Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Rosanne C. van Deuren
- Radboud University Medical Center, Department of Human Genetics, Geert Grooteplein Zuid 855, 6525 GA Nijmegen, the Netherlands
| | - Alexander Hoischen
- Radboud University Medical Center, Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA Nijmegen, the Netherlands
- Radboud University Medical Center, Department of Human Genetics, Geert Grooteplein Zuid 855, 6525 GA Nijmegen, the Netherlands
- Radboud Expertise Center for Immunodeficiency and Autoinflammation and Radboud Center for Infectious Disease (RCI), Radboud University Medical Center, Geert Grooteplein-Zuid 10, 6525 GA Nijmegen, the Netherlands
| | - Niels P. Riksen
- Radboud University Medical Center, Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA Nijmegen, the Netherlands
| | - Adrian C. Iancu
- “Iuliu Hatieganu” University of Medicine and Pharmacy, Department of Cardiology –“Niculae Stăncioiu” Heart Institute, 19-21 Calea Moților, 400001 Cluj-Napoca, Romania
| |
Collapse
|
10
|
Prater MC, Scheurell AR, Paton CM, Cooper JA. No Observed Difference in Inflammatory and Coagulation Markers Following Diets Rich in n-6 Polyunsaturated Fat vs Monounsaturated Fat in Adults With Untreated Hypercholesterolemia: A Randomized Trial. J Acad Nutr Diet 2024; 124:205-214.e1. [PMID: 37619782 DOI: 10.1016/j.jand.2023.08.127] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 08/08/2023] [Accepted: 08/17/2023] [Indexed: 08/26/2023]
Abstract
BACKGROUND Inflammatory and prothrombotic responses are hallmark to the progression of cardiovascular disease and may be influenced by the type of dietary fat. Cottonseed oil (CSO) is rich in n-6 polyunsaturated fats and improves traditional cardiovascular disease risk factors such as cholesterol profiles. However, some clinicians are still hesitant to promote n-6 polyunsaturated fats consumption despite growing evidence suggesting they may not be independently pro-inflammatory. OBJECTIVE To investigate the inflammatory and coagulation marker responses to an 8-week diet intervention rich in either CSO or olive oil (OO) (OO is rich in monounsaturated fat) in adults with untreated hypercholesterolemia. DESIGN This was a secondary analysis of a parallel-arm randomized clinical trial with the main outcome of cholesterol measures. PARTICIPANTS/SETTING Participants included in this analysis were 42 sedentary adults aged 30 to 75 years (62% women) in the Athens, GA, area, between May 2018 and June 2021, with untreated hypercholesterolemia or elevated blood lipids and body mass index >18.5. Hypercholesterolemia was defined as at least two blood lipid levels in a borderline undesirable/at risk range (total cholesterol level ≥180 mg/dL, low-density lipoprotein cholesterol level ≥110 mg/dL, high-density lipoprotein cholesterol level <50 mg/dL, or triglyceride level ≥130 mg/dL), or at least one in an undesirable range (total cholesterol level ≥240 mg/dL, low-density lipoprotein cholesterol level ≥160 mg/dL, high-density lipoprotein cholesterol level <40 mg/dL, or triglyceride level ≥200 mg/dL). INTERVENTION Participants were randomly assigned to either the CSO or OO group in a partial outpatient feeding trial. Meals from the study provided approximately 60% of their energy needs with 30% of energy needs from either CSO or OO for 8 weeks. Participants fulfilled their remaining energy needs with meals of their choosing. MAIN OUTCOME MEASURES Fasting plasma concentrations of inflammatory markers, including C-reactive protein, tumor necrosis factor-α, interleukin-6, and interleukin-1β were measured at baseline and 8 weeks. Markers of coagulation potential, including plasminogen activator inhibitor-1, and tissue factor were measured at the same time points. STATISTICAL ANALYSES PERFORMED Repeated measures linear mixed models were used with treatment and visit in the model for analyses of all biochemical markers. RESULTS There were no significant differences in fasting C-reactive protein (P = 0.70), tumor necrosis factor-α (P = 0.98), interleukin-6 (P = 0.21), interleukin-1β (P = 0.13), plasminogen activator inhibitor-1 (P = 0.29), or tissue factor (P = 0.29) between groups across the intervention. CONCLUSIONS Inflammation and coagulation marker responses to diets rich in CSO vs OO were not significantly different between groups, and neither group showed changes in these markers in adults with untreated hypercholesterolemia. This provides additional evidence suggesting that dietary n-6 polyunsaturated fats may not promote inflammation compared with monounsaturated fatty acids, even in adults at increased risk for cardiovascular disease.
Collapse
Affiliation(s)
- M Catherine Prater
- Department of Nutritional Sciences, University of Georgia, Athens, Georgia
| | - Alexis R Scheurell
- Department of Nutritional Sciences, University of Georgia, Athens, Georgia
| | - Chad M Paton
- Department of Nutritional Sciences, University of Georgia, Athens, Georgia; Department of Food Science and Technology, University of Georgia, Athens, Georgia
| | - Jamie A Cooper
- Department of Kinesiology, University of Georgia, Athens, Georgia.
| |
Collapse
|
11
|
Martinelli N, Moruzzi S, Udali S, Castagna A, Di Santo L, Ambrosani F, Baroni M, Pattini P, Pizzolo F, Ruzzenente A, Conci S, Grusse M, Campagnaro T, Van Dreden P, Guglielmi A, Bernardi F, Olivieri O, Friso S. Tissue factor pathway-related biomarkers in liver cancer: activated factor VII-antithrombin complex and tissue factor mRNA levels are associated with mortality. Res Pract Thromb Haemost 2024; 8:102310. [PMID: 38282902 PMCID: PMC10818084 DOI: 10.1016/j.rpth.2023.102310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 12/06/2023] [Accepted: 12/15/2023] [Indexed: 01/30/2024] Open
Abstract
Background Tissue factor (TF), the main initiator of the coagulation cascade, plays a role in cancer progression and prognosis. Activated factor VII-antithrombin complex (FVIIa-AT) is considered an indirect marker of TF exposure by reflecting TF-FVIIa interaction. Objectives To assess the link between FVIIa-AT plasma levels, TF messenger RNA (mRNA) expression, and survival in cancer. Methods TF pathway-related coagulation biomarkers were assessed in 136 patients with cancer (52 with hepatocellular carcinoma, 41 with cholangiocarcinoma, and 43 with colon cancer) undergoing surgical intervention with curative intent. TF mRNA expression analysis in neoplastic vs nonneoplastic liver tissues was evaluated in a subgroup of 91 patients with primary liver cancer. Results FVIIa-AT levels were higher in patients with cancer than in 136 sex- and age-matched cancer-free controls. In patients with cancer, high levels of FVIIa-AT and total TF pathway inhibitor were associated with an increased mortality risk after adjustment for confounders, but only FVIIa-AT remained a predictor of mortality by including both FVIIa-AT and total TF pathway inhibitor in Cox regression (hazard ratio, 2.80; 95% CI, 1.23-6.39; the highest vs the lowest quartile). This association remained significant even after adjustment for extracellular vesicle-associated TF-dependent procoagulant activity. In the subgroup of patients with primary liver cancer, patients with high TF mRNA levels had an increased mortality risk compared with that for those with low TF mRNA levels (hazard ratio, 1.92; 95% CI, 1.03-3.57), and there was a consistent correlation among high FVIIa-AT levels, high TF mRNA levels, and increased risk of mortality. Conclusion High FVIIa-AT levels may allow the identification of patients with cancer involving high TF expression and predict a higher mortality risk in liver cancer.
Collapse
Affiliation(s)
| | - Sara Moruzzi
- Department of Medicine, University of Verona, Verona, Italy
| | - Silvia Udali
- Department of Medicine, University of Verona, Verona, Italy
| | | | - Laura Di Santo
- Department of Medicine, University of Verona, Verona, Italy
| | | | - Marcello Baroni
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | | | | | | | - Simone Conci
- Department of Surgery, University of Verona, Verona, Italy
| | - Matthieu Grusse
- Clinical Research Department, Diagnostica Stago, Gennevilliers, France
| | | | | | | | - Francesco Bernardi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | | | | |
Collapse
|
12
|
Heidari Z, Naeimzadeh Y, Fallahi J, Savardashtaki A, Razban V, Khajeh S. The Role of Tissue Factor In Signaling Pathways of Pathological Conditions and Angiogenesis. Curr Mol Med 2024; 24:1135-1151. [PMID: 37817529 DOI: 10.2174/0115665240258746230919165935] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/10/2023] [Accepted: 07/27/2023] [Indexed: 10/12/2023]
Abstract
Tissue factor (TF) is an integral transmembrane protein associated with the extrinsic coagulation pathway. TF gene expression is regulated in response to inflammatory cytokines, bacterial lipopolysaccharides, and mechanical injuries. TF activity may be affected by phosphorylation of its cytoplasmic domain and alternative splicing. TF acts as the primary initiator of physiological hemostasis, which prevents local bleeding at the injury site. However, aberrant expression of TF, accompanied by the severity of diseases and infections under various pathological conditions, triggers multiple signaling pathways that support thrombosis, angiogenesis, inflammation, and metastasis. Protease-activated receptors (PARs) are central in the downstream signaling pathways of TF. In this study, we have reviewed the TF signaling pathways in different pathological conditions, such as wound injury, asthma, cardiovascular diseases (CVDs), viral infections, cancer and pathological angiogenesis. Angiogenic activities of TF are critical in the repair of wound injuries and aggressive behavior of tumors, which are mainly performed by the actions of vascular endothelial growth factor (VEGF) and hypoxia-inducible factor-1 (HIF1-α). Pro-inflammatory effects of TF have been reported in asthma, CVDs and viral infections, including COVID-19, which result in tissue hypertrophy, inflammation, and thrombosis. TF-FVII induces angiogenesis via clotting-dependent and -independent mechanisms. Clottingdependent angiogenesis is induced via the generation of thrombin and cross-linked fibrin network, which facilitate vessel infiltration and also act as a reservoir for endothelial cells (ECs) growth factors. Expression of TF in tumor cells and ECs triggers clotting-independent angiogenesis through induction of VEGF, urokinase-type plasminogen activator (uPAR), early growth response 1 (EGR1), IL8, and cysteine-rich angiogenic inducer 61 (Cyr61).
Collapse
Affiliation(s)
- Zahra Heidari
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Yasaman Naeimzadeh
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jafar Fallahi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Vahid Razban
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sahar Khajeh
- Bone and Joint Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
13
|
Mazzarino M, Cetin E, Bartosova M, Marinovic I, Ipseiz N, Hughes TR, Schmitt CP, Ramji DP, Labéta MO, Raby AC. Therapeutic targeting of chronic kidney disease-associated DAMPs differentially contributing to vascular pathology. Front Immunol 2023; 14:1240679. [PMID: 37849759 PMCID: PMC10577224 DOI: 10.3389/fimmu.2023.1240679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/08/2023] [Indexed: 10/19/2023] Open
Abstract
Chronic Kidney Disease (CKD) is associated with markedly increased cardiovascular (CV) morbidity and mortality. Chronic inflammation, a hallmark of both CKD and CV diseases (CVD), is believed to drive this association. Pro-inflammatory endogenous TLR agonists, Damage-Associated Molecular Patterns (DAMPs), have been found elevated in CKD patients' plasma and suggested to promote CVD, however, confirmation of their involvement, the underlying mechanism(s), the extent to which individual DAMPs contribute to vascular pathology in CKD and the evaluation of potential therapeutic strategies, have remained largely undescribed. A multi-TLR inhibitor, soluble TLR2, abrogated chronic vascular inflammatory responses and the increased aortic atherosclerosis-associated gene expression observed in nephropathic mice, without compromising infection clearance. Mechanistically, we confirmed elevation of 4 TLR DAMPs in CKD patients' plasma, namely Hsp70, Hyaluronic acid, HMGB-1 and Calprotectin, which displayed different abilities to promote key cellular responses associated with vascular inflammation and progression of atherosclerosis in a TLR-dependent manner. These included loss of trans-endothelial resistance, enhanced monocyte migration, increased cytokine production, and foam cell formation by macrophages, the latter via cholesterol efflux inhibition. Calprotectin and Hsp70 most consistently affected these functions. Calprotectin was further elevated in CVD-diagnosed CKD patients and strongly correlated with the predictor of CV events CRP. In nephropathic mice, Calprotectin blockade robustly reduced vascular chronic inflammatory responses and pro-atherosclerotic gene expression in the blood and aorta. Taken together, these findings demonstrated the critical extent to which the DAMP-TLR pathway contributes to vascular inflammatory and atherogenic responses in CKD, revealed the mechanistic contribution of specific DAMPs and described two alternatives therapeutic approaches to reduce chronic vascular inflammation and lower CV pathology in CKD.
Collapse
Affiliation(s)
- Morgane Mazzarino
- Division of Infection & Immunity, Cardiff University, Cardiff, United Kingdom
- Wales Kidney Research Unit, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Esra Cetin
- Division of Infection & Immunity, Cardiff University, Cardiff, United Kingdom
- Wales Kidney Research Unit, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Maria Bartosova
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Iva Marinovic
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Natacha Ipseiz
- Division of Infection & Immunity, Cardiff University, Cardiff, United Kingdom
| | - Timothy R. Hughes
- Division of Infection & Immunity, Cardiff University, Cardiff, United Kingdom
| | - Claus Peter Schmitt
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Dipak P. Ramji
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Mario O. Labéta
- Division of Infection & Immunity, Cardiff University, Cardiff, United Kingdom
- Wales Kidney Research Unit, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Anne-Catherine Raby
- Division of Infection & Immunity, Cardiff University, Cardiff, United Kingdom
- Wales Kidney Research Unit, School of Medicine, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
14
|
Yuan F, Li S, Huang B, Hu Y, Zeng X, Peng Y, Du C, Rong M. Molecular mechanism by which spider-driving peptide potentiates coagulation factors. Biomed Pharmacother 2023; 166:115421. [PMID: 37660649 DOI: 10.1016/j.biopha.2023.115421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/05/2023] Open
Abstract
Hemostasis is a crucial process that quickly forms clots at injury sites to prevent bleeding and infections. Dysfunctions in this process can lead to hemorrhagic disorders, such as hemophilia and thrombocytopenia purpura. While hemostatic agents are used in clinical treatments, there is still limited knowledge about potentiators targeting coagulation factors. Recently, LCTx-F2, a procoagulant spider-derived peptide, was discovered. This study employed various methods, including chromogenic substrate analysis and dynamic simulation, to investigate how LCTx-F2 enhances the activity of thrombin and FXIIa. Our findings revealed that LCTx-F2 binds to thrombin and FXIIa in a similar manner, with the N-terminal penetrating the active-site cleft of the enzymes and the intermediate section reinforcing the peptide-enzyme connection. Interestingly, the C-terminal remained at a considerable distance from the enzymes, as evidenced by the retention of affinity for both enzymes using truncated peptide T-F2. Furthermore, results indicated differences in the bonding relationship of critical residues between thrombin and FXIIa, with His13 facilitating binding to thrombin and Arg7 being required for binding to FXIIa. Overall, our study sheds light on the molecular mechanism by which LCTx-F2 potentiates coagulation factors, providing valuable insights that may assist in designing drugs targeting procoagulation factors.
Collapse
Affiliation(s)
- Fuchu Yuan
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410006, China
| | - Shuwan Li
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410006, China
| | - Biao Huang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410006, China
| | - Ya Hu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410006, China
| | - Xiongzhi Zeng
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410006, China
| | - Yanmei Peng
- Institute of Innovative Medicine, Hunan Academy of Chinese Medicine, Changsha, Hunan 410006, China
| | - Canwei Du
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, Hunan, China; Institute of Innovative Medicine, Hunan Academy of Chinese Medicine, Changsha, Hunan 410006, China.
| | - Mingqiang Rong
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410006, China.
| |
Collapse
|
15
|
Bonetti NR, Jouppila AS, Saeedi Saravi SS, Cooley BC, Pasterk L, Liberale LL, Gobbato S, Lüscher TF, Camici GG, Lassila RP, Beer JH. Intravenously administered APAC, a dual AntiPlatelet AntiCoagulant, targets arterial injury site to inhibit platelet thrombus formation and tissue factor activity in mice. Thromb Res 2023; 228:163-171. [PMID: 37331119 DOI: 10.1016/j.thromres.2023.04.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/21/2023] [Accepted: 04/11/2023] [Indexed: 06/20/2023]
Abstract
INTRODUCTION Arterial thrombosis is the main underlying mechanism of acute atherothrombosis. Combined antiplatelet and anticoagulant regimens prevent thrombosis but increase bleeding rates. Mast cell-derived heparin proteoglycans have local antithrombotic properties, and their semisynthetic dual AntiPlatelet and AntiCoagulant (APAC) mimetic may provide a new efficacious and safe tool for arterial thrombosis. We investigated the in vivo impact of intravenous APAC (0.3-0.5 mg/kg; doses chosen according to pharmacokinetic studies) in two mouse models of arterial thrombosis and the in vitro actions in mouse platelets and plasma. MATERIALS AND METHODS Platelet function and coagulation were studied with light transmission aggregometry and clotting times. Carotid arterial thrombosis was induced either by photochemical injury or surgically exposing vascular collagen after infusion of APAC, UFH or vehicle. Time to occlusion, targeting of APAC to the vascular injury site and platelet deposition on these sites were assessed by intra-vital imaging. Tissue factor activity (TF) of the carotid artery and in plasma was captured. RESULTS APAC inhibited platelet responsiveness to agonist stimulation (collagen and ADP) and prolonged APTT and thrombin time. After photochemical carotid injury, APAC-treatment prolonged times to occlusion in comparison with UFH or vehicle, and decreased TF both in carotid lysates and plasma. Upon binding from circulation to vascular collagen-exposing injury sites, APAC reduced the in situ platelet deposition. CONCLUSIONS Intravenous APAC targets arterial injury sites to exert local dual antiplatelet and anticoagulant actions and attenuates thrombosis upon carotid injuries in mice. Systemic APAC provides local efficacy, highlighting APAC as a novel antithrombotic to reduce cardiovascular complications.
Collapse
Affiliation(s)
- Nicole R Bonetti
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Department of Internal Medicine, Cantonal Hospital Baden, Switzerland
| | - Annukka S Jouppila
- Helsinki University Hospital Clinical Research Institute, Helsinki, Finland
| | - Seyed Soheil Saeedi Saravi
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Brian C Cooley
- Department of Pathology and Laboratory Medicine, Animal Surgery Core Lab, McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Lisa Pasterk
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Luca L Liberale
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Sara Gobbato
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Department of Internal Medicine, Cantonal Hospital Baden, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Royal Brompton and Harefield Hospital Trusts and National Heart and Lung Institute, Imperial College, London, UK
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; University Heart Center, University Hospital Zurich, Switzerland; Department of Research and Education, University Hospital Zurich, Switzerland
| | - Riitta P Lassila
- Coagulation Disorders Unit, University of Helsinki and Departments of Hematology and Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland; Helsinki University, Faculty of Medicine, Research Program in Systems Oncology, Helsinki, Finland; Aplagon Ltd., Helsinki, Finland.
| | - Jürg H Beer
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Department of Internal Medicine, Cantonal Hospital Baden, Switzerland
| |
Collapse
|
16
|
Sobolevskaya EV, Shumkov OA, Smagin MA, Guskov AE, Malysheva AV, Atuchin VV, Nimaev VV. Markers of Restenosis after Percutaneous Transluminal Balloon Angioplasty in Patients with Critical Limb Ischemia. Int J Mol Sci 2023; 24:ijms24109096. [PMID: 37240440 DOI: 10.3390/ijms24109096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/14/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Among cardiovascular diseases, chronic obliterating lesions of the arteries of lower extremities, which are one of the important problems of modern healthcare, are distinguished. In most cases, the cause of damage to the arteries of lower extremities is atherosclerosis. The most severe form is chronic ischemia, characterized by pain at rest and ischemic ulcers, ultimately increasing the risk of limb loss and cardiovascular mortality. Therefore, patients with critical limb ischemia need limb revascularization. Percutaneous transluminal balloon angioplasty is one of the least invasive and safe approaches, with advantages for patients with comorbidities. However, after this procedure, restenosis is still possible. Early detection of changes in the composition of some molecules as markers of restenosis will help screen patients at the risk of restenosis, as well as find ways to apply efforts for further directions of inhibition of this process. The purpose of this review is to provide the most important and up-to-date information on the mechanisms of restenosis development, as well as possible predictors of their occurrence. The information collected in this publication may be useful in predicting outcomes after surgical treatment and will also find new ways for the target implication to the mechanisms of development of restenosis and atherosclerosis.
Collapse
Affiliation(s)
- Elvira V Sobolevskaya
- Laboratory of Surgical Lymphology and Lymph-Detoxication, Research Institute of Clinical and Experimental Lymphology-Branch of the Institute of Cytology and Genetics, SB RAS, Novosibirsk 630117, Russia
| | - Oleg A Shumkov
- Laboratory of Surgical Lymphology and Lymph-Detoxication, Research Institute of Clinical and Experimental Lymphology-Branch of the Institute of Cytology and Genetics, SB RAS, Novosibirsk 630117, Russia
| | - Mikhail A Smagin
- Laboratory of Surgical Lymphology and Lymph-Detoxication, Research Institute of Clinical and Experimental Lymphology-Branch of the Institute of Cytology and Genetics, SB RAS, Novosibirsk 630117, Russia
| | - Andrey E Guskov
- Laboratory of Scientometrics and Scientific Communications, Russian Research Institute of Economics, Politics and Law in Science and Technology, Moscow 127254, Russia
| | - Alexandra V Malysheva
- Laboratory of Scientometrics and Scientific Communications, Russian Research Institute of Economics, Politics and Law in Science and Technology, Moscow 127254, Russia
| | - Victor V Atuchin
- Laboratory of Optical Materials and Structures, Institute of Semiconductor Physics, SB RAS, Novosibirsk 630090, Russia
- Research and Development Department, Kemerovo State University, Kemerovo 650000, Russia
- Department of Industrial Machinery Design, Novosibirsk State Technical University, Novosibirsk 630073, Russia
- R&D Center "Advanced Electronic Technologies", Tomsk State University, Tomsk 634034, Russia
| | - Vadim V Nimaev
- Laboratory of Surgical Lymphology and Lymph-Detoxication, Research Institute of Clinical and Experimental Lymphology-Branch of the Institute of Cytology and Genetics, SB RAS, Novosibirsk 630117, Russia
| |
Collapse
|
17
|
Rolling CC, Barrett TJ, Berger JS. Platelet-monocyte aggregates: molecular mediators of thromboinflammation. Front Cardiovasc Med 2023; 10:960398. [PMID: 37255704 PMCID: PMC10225702 DOI: 10.3389/fcvm.2023.960398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 04/24/2023] [Indexed: 06/01/2023] Open
Abstract
Platelets, key facilitators of primary hemostasis and thrombosis, have emerged as crucial cellular mediators of innate immunity and inflammation. Exemplified by their ability to alter the phenotype and function of monocytes, activated platelets bind to circulating monocytes to form monocyte-platelet aggregates (MPA). The platelet-monocyte axis has emerged as a key mechanism connecting thrombosis and inflammation. MPA are elevated across the spectrum of inflammatory and autoimmune disorders, including cardiovascular disease, systemic lupus erythematosus (SLE), and COVID-19, and are positively associated with disease severity. These clinical disorders are all characterized by an increased risk of thromboembolic complications. Intriguingly, monocytes in contact with platelets become proinflammatory and procoagulant, highlighting that this interaction is a central element of thromboinflammation.
Collapse
Affiliation(s)
- Christina C. Rolling
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
- Department of Oncology and Hematology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tessa J. Barrett
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| | - Jeffrey S. Berger
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
18
|
Liberale L, Kraler S, Puspitasari YM, Bonetti NR, Akhmedov A, Ministrini S, Montecucco F, Marx N, Lehrke M, Hartmann NUK, Beer JH, Wenzl FA, Paneni F, Lüscher TF, Camici GG. SGLT-2 inhibition by empagliflozin has no effect on experimental arterial thrombosis in a murine model of low-grade inflammation. Cardiovasc Res 2023; 119:843-856. [PMID: 35993135 DOI: 10.1093/cvr/cvac126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/04/2022] [Indexed: 11/14/2022] Open
Abstract
AIMS Low-grade inflammation couples dysmetabolic states to insulin resistance and atherosclerotic cardiovascular (CV) disease (ASCVD). Selective sodium-glucose co-transporter 2 (SGLT-2) inhibition by empagliflozin improves clinical outcomes in patients with ASCVD independently of its glucose lowering effects. Yet, its mechanism of action remains largely undetermined. Here, we aimed to test whether empagliflozin affects arterial thrombus formation in baseline (BSL) conditions or low-grade inflammatory states, a systemic milieu shared among patients with ASCVD. METHODS AND RESULTS Sixteen-week-old C57BL/6 mice were randomly assigned to acute administration of empagliflozin (25 mg/kg body weight) or vehicle, of which a subgroup was pre-treated biweekly over 4 weeks with super-low-dose lipopolysaccharide (LPS; 5 ng/kg body weight), before carotid thrombosis was induced by photochemical injury. The between-group difference in Doppler-flow probe detected time-to-occlusion remained within the predefined equivalence margin (Δ = |10.50|), irrespective of low-grade inflammation (95% confidence interval, -9.82 to 8.85 and -9.20 to 9.69), while glucose dropped by 1.64 and 4.84 mmoL/L, respectively. Ex vivo platelet aggregometry suggested similar activation status, corroborated by unchanged circulating platelet-factor 4 plasma levels. In concert, carotid PAI-1 expression and tissue factor (TF) activity remained unaltered upon SGLT-2 inhibition, and no difference in plasma D-dimer levels was detected, suggesting comparable coagulation cascade activation and fibrinolytic activity. In human aortic endothelial cells pre-treated with LPS, empagliflozin neither changed TF activity nor PAI-1 expression. Accordingly, among patients with established ASCVD or at high CV risk randomized to a daily dose of 10 mg empagliflozin signatures of thrombotic (i.e. TF) and fibrinolytic activity (i.e. PAI-1) remained unchanged, while plasma glucose declined significantly during 3 months of follow-up. CONCLUSION SGLT-2 inhibition by empagliflozin does not impact experimental arterial thrombus formation, neither under BSL conditions nor during sustained low-grade inflammation, and has no impact on proxies of thrombotic/fibrinolytic activity in patients with ASCVD. The beneficial pleiotropic effects of empagliflozin are likely independent of pathways mediating arterial thrombosis.
Collapse
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
| | - Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Yustina M Puspitasari
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Nicole R Bonetti
- University Heart Center, Department of Cardiology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Alexander Akhmedov
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- Internal Medicine, Angiology and Atherosclerosis, Department of Medicine and Surgery, University of Perugia, 06123 Perugia, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, 16132 Genoa, Italy
| | - Nikolaus Marx
- Department of Internal Medicine I, University Hospital Aachen, RWTH, 52074 Aachen, Germany
| | - Michael Lehrke
- Department of Internal Medicine I, University Hospital Aachen, RWTH, 52074 Aachen, Germany
| | - Niels-Ulrik K Hartmann
- Department of Internal Medicine I, University Hospital Aachen, RWTH, 52074 Aachen, Germany
| | - Jürg H Beer
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- Department of Internal Medicine, Cantonal Hospital of Baden, 5404 Baden, Switzerland
| | - Florian A Wenzl
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Francesco Paneni
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- University Heart Center, Department of Cardiology, University Hospital Zurich, 8091 Zurich, Switzerland
- Department of Research and Education, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- Royal Brompton and Harefield Hospitals and Imperial College, SW3 6NP London, UK
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- University Heart Center, Department of Cardiology, University Hospital Zurich, 8091 Zurich, Switzerland
- Department of Research and Education, University Hospital Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
19
|
Iannucci J, Grammas P. Thrombin, a Key Driver of Pathological Inflammation in the Brain. Cells 2023; 12:cells12091222. [PMID: 37174621 PMCID: PMC10177239 DOI: 10.3390/cells12091222] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/21/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Neurodegenerative diseases, including Alzheimer's disease (AD), are major contributors to death and disability worldwide. A multitude of evidence suggests that neuroinflammation is critical in neurodegenerative disease processes. Exploring the key mediators of neuroinflammation in AD, a prototypical neurodegenerative disease, could help identify pathologic inflammatory mediators and mechanisms in other neurodegenerative diseases. Elevated levels of the multifunctional inflammatory protein thrombin are commonly found in conditions that increase AD risk, including diabetes, atherosclerosis, and traumatic brain injury. Thrombin, a main driver of the coagulation cascade, has been identified as important to pathological events in AD and other neurodegenerative diseases. Furthermore, recent evidence suggests that coagulation cascade-associated proteins act as drivers of inflammation in the AD brain, and studies in both human populations and animal models support the view that abnormalities in thrombin generation promote AD pathology. Thrombin drives neuroinflammation through its pro-inflammatory activation of microglia, astrocytes, and endothelial cells. Due to the wide-ranging pro-inflammatory effects of thrombin in the brain, inhibiting thrombin could be an effective strategy for interrupting the inflammatory cascade which contributes to neurodegenerative disease progression and, as such, may be a potential therapeutic target for AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Jaclyn Iannucci
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | | |
Collapse
|
20
|
Gao Q, Yang L, Teng F, Cheng XY, Guo SB. The tissue factor expression on CD14++CD16-monocytes is a new markers in the Chinese Han older adult population with sepsis: A prospective study. Exp Gerontol 2023; 171:112012. [PMID: 36347361 DOI: 10.1016/j.exger.2022.112012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 10/27/2022] [Accepted: 10/31/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVE To investigate the tissue factor (TF) expression on different subsets of monocyte and tissue factor secretion of peripheral blood and evaluate their association with the prognosis of sepsis in the Chinese older adult population. METHODS Monocyte subsets and TF expression on different subsets of monocyte were measured using flow cytometry in 80 older adult sepsis patients and 40 age and sex matched healthy controls. Plasma level of TF was measured using ELISA (enzyme-linked immunosorbent assay) method. RESULTS TF expression on CD14++CD16- (MO1) monocyte was lower in death (28-day non-survivor) group compared with survival (28-day survivor) groups [1.01 % (0.58 %, 1.62 %) vs. 3.66 % (1.32 %, 6.93 %), p = 0.001]. The plasma level of TF was increased in death group compared to survival group according to the 28-day mortality [109.2 (67.3, 154.2) vs. 62.1 (44.7, 115.5) pg/mL, p = 0.031]. Logistic regression analysis showed TF expression on MO1 monocyte (β = -0.776, OR = 0.460, CI: 0.251, 0.843, p = 0.012) was independently associated with the 28-day mortality. The ROC (receiver operating characteristic) curve showed that the AUC (area under the curve) of the TF expression on MO1 monocyte for predicting 28-day mortality was 0.846 (p < 0.001). CONCLUSION The TF expression on CD14++CD16- monocyte is a new marker for the prognosis of older adult sepsis.
Collapse
Affiliation(s)
- Qian Gao
- Emergency Department, Beijing Shijitan Hospital, Capital Medical University, No. 10 Tieyi Road, Yangfangdian, Haidian District, Beijing 100038, China
| | - Li Yang
- Emergency Department, Beijing Shijitan Hospital, Capital Medical University, No. 10 Tieyi Road, Yangfangdian, Haidian District, Beijing 100038, China
| | - Fei Teng
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, No. 8, South Road of Worker's Stadium, Chaoyang District, Beijing 100020, China
| | - Xiao-Yan Cheng
- Institute of Analysis and Testing, Beijing Academy of Science and Technology (Beijing Center for Physical and Chemical Analysis), No. 27 Xisanhuan North Road, Beijing 100094, China
| | - Shu-Bin Guo
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, No. 8, South Road of Worker's Stadium, Chaoyang District, Beijing 100020, China.
| |
Collapse
|
21
|
Ziaka M, Exadaktylos A. The Heart Is at Risk: Understanding Stroke-Heart-Brain Interactions with Focus on Neurogenic Stress Cardiomyopathy-A Review. J Stroke 2023; 25:39-54. [PMID: 36592971 PMCID: PMC9911836 DOI: 10.5853/jos.2022.02173] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 11/04/2022] [Accepted: 11/14/2022] [Indexed: 01/04/2023] Open
Abstract
In recent years, it has been convincingly demonstrated that acute brain injury may cause severe cardiac complications-such as neurogenic stress cardiomyopathy (NSC), a specific form of takotsubo cardiomyopathy. The pathophysiology of these brain-heart interactions is complex and involves sympathetic hyperactivity, activation of the hypothalamic-pituitary-adrenal axis, as well as immune and inflammatory pathways. There have been great strides in our understanding of the axis from the brain to the heart in patients with isolated acute brain injury and more specifically in patients with stroke. On the other hand, in patients with NSC, research has mainly focused on hemodynamic dysfunction due to arrhythmias, regional wall motion abnormality, or left ventricular hypokinesia that leads to impaired cerebral perfusion pressure. Comparatively little is known about the underlying secondary and delayed cerebral complications. The aim of the present review is to describe the stroke-heart-brain axis and highlight the main pathophysiological mechanisms leading to secondary and delayed cerebral injury in patients with concurrent hemorrhagic or ischemic stroke and NSC as well as to identify further areas of research that could potentially improve outcomes in this specific patient population.
Collapse
Affiliation(s)
- Mairi Ziaka
- Department of Internal Medicine, Thun General Hospital, Thun, Switzerland
| | - Aristomenis Exadaktylos
- Department of Emergency Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
22
|
Bozkaya TA, Ateş Ş, Üstündağ ÜV, Çakıcı Ç, Keskin İ, Yiğit P, Yiğitbaşı A, Emekli N. Tissue Factor and Vascular Endothelial Growth Factor in Detecting Thromboembolic Complications in Diabetic Atherosclerotic Patients. Clin Med Insights Circ Respir Pulm Med 2023; 17:11795484231167737. [PMID: 37113616 PMCID: PMC10126798 DOI: 10.1177/11795484231167737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 03/14/2023] [Indexed: 04/29/2023] Open
Abstract
Background Atherosclerosis, which is one of the leading causes of death all over the world, can create major or minor thromboembolic complications with the exponentially increasing diabetic status. Despite all the studies, the mechanism by which endothelial damage in atherosclerosis is triggered in diabetic setting is still not fully understood. Methods In this study, tissue factor (TF), which is thought to act together in the formation of vasular endothelial growth factor (VEGF-A) and coagulopathy in diabetic atherosclerotic patients, may be an important indicator in this regard, a total of 100 cases who were undergone off-pump coronary artery bypass (OPCAB) which were at same risk group examined by dividing into diabetic status. Early postoperative process and biochemical parameters analyzed in terms of TF and VEGF-A levels measured before and after the operation. Results TF and VEGF-A expression of the T1DM group were statistically high compared to non-diabetics. Significantly longer hospital stays with changes in TF and VEGF-A were found in patients in the diabetic group compared to pre- and postoperatively, respectively; TF (95% CI: 0.879-0.992; p = 0.025), VEGF-A (95% CI: 0.964-0.991; p = 0.001) and hospital stay (95% CI: 1.96-7.49; p = 0.0001). Preoperatively measured carotid intima-media thickness (CT) was higher in diabetics and was significantly associated with atrial fibrillation (AF), (r = 0.873). Surgical team and protocols were same and OPCAB procedures were routinely applied to all patients in our clinic. No minor or major events were observed in any of the cases. Conclusion TF and VEGF-A values in patients with diabetic atherosclerosis may be important in the early detection of thromboembolic complications.
Collapse
Affiliation(s)
- Tijen Alkan Bozkaya
- Department of Cardiovascular Surgery, Koç University Hospital, Istanbul, Turkey
- Department of Cardiovascular Surgery, Yeditepe University Hospital, Istanbul, Turkey
- Biochemistry Program, Institute of Health Sciences, İstanbul Medipol University, Istanbul, Turkey
- Tijen Alkan Bozkaya, Department of Cardiovascular Surgery, Yeditepe University Hospital, Istanbul, Turkey.
| | - Şanser Ateş
- Department of Cardiovascular Surgery, Koç University Hospital, Istanbul, Turkey
| | - Ünsal Veli Üstündağ
- Departmant of Biochemistry, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Çağrı Çakıcı
- Departmant of Biochemistry, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - İlknur Keskin
- Department of Histology and Embryology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Pakize Yiğit
- Department of Biostatistics and Medical Informatics, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Ahmet Yiğitbaşı
- Biochemistry Program, Institute of Health Sciences, İstanbul Medipol University, Istanbul, Turkey
| | - Nesrin Emekli
- Departmant of Biochemistry, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| |
Collapse
|
23
|
Martinelli N, Rigoni AM, De Marchi S, Osti N, Donini M, Montagnana M, Castagna A, Pattini P, Udali S, De Franceschi L, Tinazzi E, Mazzi F, Moruzzi S, Argentino G, Delfino L, Sartori G, Azzini AM, Tacconelli E, Van Dreden P, Lippi G, Girelli D, Olivieri O, Friso S, Pizzolo F. High Plasma Levels of Activated Factor VII-Antithrombin Complex Point to Increased Tissue Factor Expression in Patients with SARS-CoV-2 Pneumonia: A Potential Link with COVID-19 Prothrombotic Diathesis. Diagnostics (Basel) 2022; 12:diagnostics12112792. [PMID: 36428852 PMCID: PMC9689539 DOI: 10.3390/diagnostics12112792] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/04/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causal agent of coronavirus disease 2019 (COVID-19), in which coagulation abnormalities and endothelial dysfunction play a key pathogenic role. Tissue factor (TF) expression is triggered by endothelial dysfunction. Activated factor VII-antithrombin (FVIIa-AT) complex reflects indirectly FVIIa-TF interaction and has been proposed as a potential biomarker of prothrombotic diathesis. FVIIa-AT plasma concentration was measured in 40 patients (30 males and 10 females; 64.8 ± 12.3 years) admitted with SARS-CoV-2 pneumonia during the first pandemic wave in Italy. Two sex- and age-matched cohorts without COVID-19, with or without signs of systemic inflammation, were used to compare FVIIa-AT data. The FVIIa-AT plasma levels in COVID-19 patients were higher than those in non-COVID-19 subjects, either with or without inflammation, while no difference was observed among non-COVID-19 subjects. The association between COVID-19 and FVIIa-AT levels remained significant after adjustment for sex, age, C-reactive protein, renal function, fibrinogen, prothrombin time and activated partial thromboplastin time. Our results indicate that SARS-CoV-2 infection, at least during the first pandemic wave, was characterized by high FVIIa-AT levels, which may suggest an enhanced FVIIa-TF interaction in COVID-19, potentially consistent with SARS-CoV-2-induced endotheliopathy.
Collapse
Affiliation(s)
- Nicola Martinelli
- Department of Medicine, University of Verona, 37129 Verona, Italy
- Correspondence: ; Tel.: +39-045-8126658; Fax: +39-045-8027473
| | - Anna Maria Rigoni
- Angiology Unit, Department of Cardiovascular and Thoracic, Azienda Ospedaliera Universitaria Integrata, 37126 Verona, Italy
| | - Sergio De Marchi
- Department of Medicine, University of Verona, 37129 Verona, Italy
| | - Nicola Osti
- Department of Medicine, University of Verona, 37129 Verona, Italy
| | - Martino Donini
- Department of Medicine, University of Verona, 37129 Verona, Italy
| | - Martina Montagnana
- Section of Clinical Biochemistry, University of Verona, 37129 Verona, Italy
| | | | - Patrizia Pattini
- Department of Medicine, University of Verona, 37129 Verona, Italy
| | - Silvia Udali
- Department of Medicine, University of Verona, 37129 Verona, Italy
| | | | - Elisa Tinazzi
- Department of Medicine, University of Verona, 37129 Verona, Italy
| | - Filippo Mazzi
- Department of Medicine, University of Verona, 37129 Verona, Italy
| | - Sara Moruzzi
- Department of Medicine, University of Verona, 37129 Verona, Italy
| | | | - Lorenzo Delfino
- Department of Medicine, University of Verona, 37129 Verona, Italy
| | - Giulia Sartori
- Department of Medicine, University of Verona, 37129 Verona, Italy
| | - Anna Maria Azzini
- Department of Diagnostics and Public Health, University of Verona, 37129 Verona, Italy
| | - Evelina Tacconelli
- Department of Diagnostics and Public Health, University of Verona, 37129 Verona, Italy
| | - Patrick Van Dreden
- Clinical Research Department, Diagnostica Stago, 92230 Gennevilliers, France
| | - Giuseppe Lippi
- Section of Clinical Biochemistry, University of Verona, 37129 Verona, Italy
| | - Domenico Girelli
- Department of Medicine, University of Verona, 37129 Verona, Italy
| | | | - Simonetta Friso
- Department of Medicine, University of Verona, 37129 Verona, Italy
| | | |
Collapse
|
24
|
Yokoi M, Ito T, Yamamoto J, Mori K, Nakasuka K, Fujita H, Kitada S, Goto T, Seo Y. A Case of Intraprocedural Thrombotic Events During Percutaneous Coronary Intervention Due to Acquired Antithrombin Deficiency-related Heparin Resistance Successfully Treated with Antithrombin Gamma Supplementation. Intern Med 2022. [PMID: 36351584 DOI: 10.2169/internalmedicine.0720-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
A 75-year-old man receiving treatment for necrotizing pancreatitis developed septic disseminated intravascular coagulation and acute coronary syndrome (ACS). During percutaneous coronary intervention (PCI), a large amount of fresh thrombi appeared after balloon dilatation for the ACS-culprit lesion. Given the low plasma AT activity and poorly prolonged activated clotting time (ACT), we suspected that acquired AT deficiency-related heparin resistance (HR) was responsible for the thrombus formation. Administration of AT gamma markedly improved ACT, and we successfully completed PCI. We suggest that AT gamma be considered a treatment option for AT deficiency-related HR and subsequent intraprocedural thrombotic events.
Collapse
Affiliation(s)
- Masashi Yokoi
- Department of Cardiology, Nagoya City University Graduate School of Medical Sciences, Japan
| | - Tsuyoshi Ito
- Department of Cardiology, Nagoya City University Graduate School of Medical Sciences, Japan
| | - Junki Yamamoto
- Department of Cardiology, Nagoya City University Graduate School of Medical Sciences, Japan
| | - Kento Mori
- Department of Cardiology, Nagoya City University Graduate School of Medical Sciences, Japan
| | - Kosuke Nakasuka
- Department of Cardiology, Nagoya City University Graduate School of Medical Sciences, Japan
| | - Hiroshi Fujita
- Department of Cardiology, Nagoya City University Graduate School of Medical Sciences, Japan
| | - Shuichi Kitada
- Department of Cardiology, Nagoya City University Graduate School of Medical Sciences, Japan
| | - Toshihiko Goto
- Department of Cardiology, Nagoya City University Graduate School of Medical Sciences, Japan
| | - Yoshihiro Seo
- Department of Cardiology, Nagoya City University Graduate School of Medical Sciences, Japan
| |
Collapse
|
25
|
Shi J, Tang Y, Liang F, Liu L, Liang N, Yang X, Zhang N, Yi Z, Zhong Y, Wang W, Zhao K. NLRP3 inflammasome contributes to endotoxin-induced coagulation. Thromb Res 2022; 214:8-15. [PMID: 35421682 DOI: 10.1016/j.thromres.2022.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/30/2022] [Accepted: 04/04/2022] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Excessive activation of the coagulation cascades leads to life-threatening disseminated intravascular coagulation (DIC) in sepsis. Two recent studies by our group and others have both demonstrated the noncanonical inflammasome is pivotal for the endotoxin or gram-negative bacterial-induced coagulation. Based on this, we further evaluated the function of the NLRP3 inflammasome, the most studied inflammasome, in endotoxin-induced coagulation. MATERIALS AND METHODS We established an endotoxin-induced coagulation model by intraperitoneal injection of sublethal doses of LPS in mice. Mice were sacrificed 8 h after injection and blood was collected for thrombin-antithrombin (TAT), plasminogen activator inhibitor-1 (PAI-1), prothrombin time (PT), D-dimer, IL-1β and tissue factor (TF) measurements by commercial ELISA. Lungs and livers were examined via HE staining images to determine injury scores and immunohistochemistry for TF expression and fibrin deposits. The role of NLRP3 activation was evaluated in wild-type (WT), Nlrp3-/-, Asc-/- (apoptosis-associated speck-like protein containing a CARD), Caspase-11-/- mice and 30 min after treatment with MCC950, a potent inhibitor of NLRP3. Western blotting and Q-PCR were performed to assess TF expression in the lungs and livers. To uncover the different effects of NLRP3 and Caspase-11, we also compared the time-dependent IL-1β release in LPS-treated Nlrp3-/- and Caspase-11-/- mice. Correlation analysis of TAT, PAI-1 were estimated the relationship of coagulation and release of IL-1β, as well as IL-1β and TF. RESULTS Inhibition of NLRP3 by MCC950 as well as NLRP3 or ASC deficiency decreased TAT, PAI-1, PT, D-dimer, and TF levels in blood and impaired the thrombus formation and fibrin deposition, as well as declined expression of TF in the liver and lung in endotoxin-induced coagulation but not caspase-11 deficiency. Impressively, IL-1β release is increased in LPS-treated Caspase-11-/- mice, but not in Nlrp3-/- mice. Moreover, the correlation analysis is indicated that downstream of the NLRP3 inflammasome, IL-1β expression, is positively correlated with TAT, PAI-1 and TF in blood circulation. CONCLUSIONS The NLRP3 inflammasome contributes to endotoxin-induced coagulation by promoting TF expression at least in part through the induction of IL-1β release. These findings broadened our understanding of the mechanism of coagulation and implicated a possible therapeutic strategy for preventing coagulation in sepsis.
Collapse
Affiliation(s)
- Jie Shi
- Department of General Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yiting Tang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Fang Liang
- Department of Hematology and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liping Liu
- Department of General Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ni Liang
- Department of Hematology and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xinyu Yang
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ningjie Zhang
- Department of Blood Transfusion, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhonjie Yi
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yanjun Zhong
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wenhua Wang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Kai Zhao
- Department of Hematology and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
26
|
Chaudhary PK, Kim S, Kim S. An Insight into Recent Advances on Platelet Function in Health and Disease. Int J Mol Sci 2022; 23:ijms23116022. [PMID: 35682700 PMCID: PMC9181192 DOI: 10.3390/ijms23116022] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/22/2022] [Accepted: 05/24/2022] [Indexed: 12/04/2022] Open
Abstract
Platelets play a variety of roles in vascular biology and are best recognized as primary hemostasis and thrombosis mediators. Platelets have a large number of receptors and secretory molecules that are required for platelet functionality. Upon activation, platelets release multiple substances that have the ability to influence both physiological and pathophysiological processes including inflammation, tissue regeneration and repair, cancer progression, and spreading. The involvement of platelets in the progression and seriousness of a variety of disorders other than thrombosis is still being discovered, especially in the areas of inflammation and the immunological response. This review represents an integrated summary of recent advances on the function of platelets in pathophysiology that connects hemostasis, inflammation, and immunological response in health and disease and suggests that antiplatelet treatment might be used for more than only thrombosis.
Collapse
|
27
|
Luchini A, Tidemand FG, Araya-Secchi R, Campana M, Cárdenas M, Arleth L. Structural model of tissue factor (TF) and TF-factor VIIa complex in a lipid membrane: A combined experimental and computational study. J Colloid Interface Sci 2022; 623:294-305. [PMID: 35594588 DOI: 10.1016/j.jcis.2022.04.147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/11/2022] [Accepted: 04/25/2022] [Indexed: 10/18/2022]
Abstract
Tissue factor (TF) is a membrane protein involved in blood coagulation. TF initiates a cascade of proteolytic reactions, ultimately leading to the formation of a blood clot. The first reaction consists of the binding of the coagulation factor VII and its conversion to the activated form, FVIIa. Here, we combined experimental, i.e. quartz crystal microbalance with dissipation monitoring and neutron reflectometry, and computational, i.e. molecular dynamics (MD) simulation, methods to derive a complete structural model of TF and TF/FVIIa complex in a lipid bilayer. This model shows that the TF transmembrane domain (TMD), and the flexible linker connecting the TMD to the extracellular domain (ECD), define the location of the ECD on the membrane surface. The average orientation of the ECD relative to the bilayer surface is slightly tilted towards the lipid headgroups, a conformation that we suggest is promoted by phosphatidylserine lipids, and favours the binding of FVIIa. On the other hand, the formation of the TF/FVIIa complex induces minor changes in the TF structure, and reduces the conformational freedom of both TF and FVIIA. Altogether we describe the protein-protein and protein-lipid interactions favouring blood coagulation, but also instrumental to the development of new drugs.
Collapse
Affiliation(s)
- Alessandra Luchini
- Niels Bohr Institute, University of Copenhagen, Universitetsparken 5, 2100 Copenhagen, Denmark.
| | | | - Raul Araya-Secchi
- Niels Bohr Institute, University of Copenhagen, Universitetsparken 5, 2100 Copenhagen, Denmark
| | - Mario Campana
- ISIS-STFC, Rutherford Appleton Laboratory, Chilton, Oxon OX11 0QX, United Kingdom
| | - Marité Cárdenas
- Biofilms Research Center for Biointerfaces and Department of Biomedical Science, Faculty of Health and Society, Malmö University, Per Albin Hanssons Väg 35, 21432 Malmö, Sweden
| | - Lise Arleth
- Niels Bohr Institute, University of Copenhagen, Universitetsparken 5, 2100 Copenhagen, Denmark
| |
Collapse
|
28
|
Jebari-Benslaiman S, Galicia-García U, Larrea-Sebal A, Olaetxea JR, Alloza I, Vandenbroeck K, Benito-Vicente A, Martín C. Pathophysiology of Atherosclerosis. Int J Mol Sci 2022; 23:ijms23063346. [PMID: 35328769 PMCID: PMC8954705 DOI: 10.3390/ijms23063346] [Citation(s) in RCA: 382] [Impact Index Per Article: 127.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/12/2022] [Accepted: 03/18/2022] [Indexed: 11/26/2022] Open
Abstract
Atherosclerosis is the main risk factor for cardiovascular disease (CVD), which is the leading cause of mortality worldwide. Atherosclerosis is initiated by endothelium activation and, followed by a cascade of events (accumulation of lipids, fibrous elements, and calcification), triggers the vessel narrowing and activation of inflammatory pathways. The resultant atheroma plaque, along with these processes, results in cardiovascular complications. This review focuses on the different stages of atherosclerosis development, ranging from endothelial dysfunction to plaque rupture. In addition, the post-transcriptional regulation and modulation of atheroma plaque by microRNAs and lncRNAs, the role of microbiota, and the importance of sex as a crucial risk factor in atherosclerosis are covered here in order to provide a global view of the disease.
Collapse
Affiliation(s)
- Shifa Jebari-Benslaiman
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain; (S.J.-B.); (I.A.); (K.V.)
- Biofisika Institute (UPV/EHU, CSIC), Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain; (U.G.-G.); (A.L.-S.)
| | - Unai Galicia-García
- Biofisika Institute (UPV/EHU, CSIC), Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain; (U.G.-G.); (A.L.-S.)
- Fundación Biofisika Bizkaia, Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain
| | - Asier Larrea-Sebal
- Biofisika Institute (UPV/EHU, CSIC), Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain; (U.G.-G.); (A.L.-S.)
- Fundación Biofisika Bizkaia, Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain
| | | | - Iraide Alloza
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain; (S.J.-B.); (I.A.); (K.V.)
- Inflammation & Biomarkers Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Bizkaia, Spain
| | - Koen Vandenbroeck
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain; (S.J.-B.); (I.A.); (K.V.)
- Inflammation & Biomarkers Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Bizkaia, Spain
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Bizkaia, Spain
| | - Asier Benito-Vicente
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain; (S.J.-B.); (I.A.); (K.V.)
- Biofisika Institute (UPV/EHU, CSIC), Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain; (U.G.-G.); (A.L.-S.)
- Correspondence: (A.B.-V.); (C.M.); Tel.: +34-946-01-2741 (C.M.)
| | - César Martín
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain; (S.J.-B.); (I.A.); (K.V.)
- Biofisika Institute (UPV/EHU, CSIC), Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain; (U.G.-G.); (A.L.-S.)
- Correspondence: (A.B.-V.); (C.M.); Tel.: +34-946-01-2741 (C.M.)
| |
Collapse
|
29
|
Nappi F, Giacinto O, Ellouze O, Nenna A, Avtaar Singh SS, Chello M, Bouzguenda A, Copie X. Association between COVID-19 Diagnosis and Coronary Artery Thrombosis: A Narrative Review. Biomedicines 2022; 10:702. [PMID: 35327504 PMCID: PMC8945192 DOI: 10.3390/biomedicines10030702] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/13/2022] [Accepted: 03/16/2022] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 is characterized by its severe respiratory effects. Data early on indicated an increased risk of mortality in patients with cardiovascular comorbidities. Early reports highlighted the multisystem inflammatory syndrome, cytokine storm, and thromboembolic events as part of the disease processes. The aim of this review is to assess the association between COVID-19 and its thrombotic complications, specifically related to the cardiovascular system. The role of neutrophil extracellular traps (NETs) is explored in the pathogenesis of the disease. The structure and anatomy of the virus are pivotal to its virulence in comparison to other α and β Coronaviridae (HCoV-229E, HCoV-OC43, HCoV-229E, HCoV-NL63, HCoV-OC43, and HCoV-HKU1). In particular, the host interaction and response may explain the variability of severity in patients. Angio tensin-converting enzyme 2 (ACE2) activation may be implicated in the cardiovascular and throm bogenic potential of the disease. The virus may also have direct effects on the endothelial lining affecting hemostasis and resulting in thrombosis through several mechanisms. Dipyridamole may have a therapeutic benefit in NET suppression. Therapeutic avenues should be concentrated on the different pathophysiological steps involving the virus and the host.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord de Saint-Denis (CCN), 36 Rue des Moulins Gémeaux, 93200 Saint-Denis, France
| | - Omar Giacinto
- Cardiovascular Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (O.G.); (A.N.); (M.C.)
| | - Omar Ellouze
- Department of Anesthesia, Centre Cardiologique du Nord, 93200 Saint-Denis, France; (O.E.); (A.B.)
| | - Antonio Nenna
- Cardiovascular Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (O.G.); (A.N.); (M.C.)
| | | | - Massimo Chello
- Cardiovascular Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (O.G.); (A.N.); (M.C.)
| | - Assine Bouzguenda
- Department of Anesthesia, Centre Cardiologique du Nord, 93200 Saint-Denis, France; (O.E.); (A.B.)
| | - Xavier Copie
- Department of Arrhythmology and Electrostimulation, Centre Cardiologique du Nord, 93200 Saint-Denis, France;
| |
Collapse
|
30
|
Lüscher TF, Davies A, Beer JH, Valgimigli M, Nienaber CA, Camm JA, Baumgartner I, Diener HC, Konstantinides SV. Towards personalized antithrombotic management with drugs and devices across the cardiovascular spectrum. Eur Heart J 2022; 43:940-958. [PMID: 34624084 DOI: 10.1093/eurheartj/ehab642] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 05/06/2021] [Accepted: 09/01/2021] [Indexed: 01/10/2023] Open
Abstract
Intravascular thrombus formation and embolization are among the most frequent events leading to a number of cardiovascular conditions with high morbidity and mortality. The underlying causes are stasis of the circulating blood, genetic and acquired coagulation disorders, and reduced antithrombotic or prothrombotic properties of the vascular wall (Virchow's triad). In the venous system, intravascular thrombi can cause venous thrombosis and pulmonary and even peripheral embolism including ischaemic stroke [through a patent foramen ovale (PFO)]. Thrombi in the left atrium and its appendage or ventricle form in the context of atrial fibrillation and infarction, respectively. Furthermore, thrombi can form on native or prosthetic aortic valves, within the aorta (in particular at sites of ulcers, aortic dissection, and abdominal aneurysms), and in cerebral and peripheral arteries causing stroke and critical limb ischaemia, respectively. Finally, thrombotic occlusion may occur in arteries supplying vital organs such the heart, brain, kidney, and extremities. Thrombus formation and embolization can be managed with anticoagulants and devices depending on where they form and embolize and on patient characteristics. Vitamin K antagonists are preferred in patients with mechanical valves, while novel oral anticoagulants are first choice in most other cardiovascular conditions, in particular venous thromboembolism and atrial fibrillation. As anticoagulants are associated with a risk of bleeding, devices such as occluders of a PFO or the left atrial appendage are preferred in patients with an increased bleeding risk. Platelet inhibitors such as aspirin and/or P2Y12 antagonists are preferred in the secondary prevention of coronary artery disease, stroke, and peripheral artery disease either alone or in combination depending on the clinical condition. A differential and personalized use of anticoagulants, platelet inhibitors, and devices is recommended and reviewed in this article.
Collapse
Affiliation(s)
- Thomas F Lüscher
- Royal Brompton & Harefield Hospitals, Heart Division, Guy Scadding Building, Dovehouse Street, Imperial College, London SW3 6LY, UK.,Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Allan Davies
- Royal Brompton & Harefield Hospitals, Heart Division, Guy Scadding Building, Dovehouse Street, Imperial College, London SW3 6LY, UK
| | - Juerg H Beer
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Marco Valgimigli
- CardioCentro, Lugano, Switzerland.,University of Bern, Bern, Switzerland
| | - Christoph A Nienaber
- Royal Brompton & Harefield Hospitals, Heart Division, Guy Scadding Building, Dovehouse Street, Imperial College, London SW3 6LY, UK
| | - John A Camm
- St. Georges University and Imperial College, London, UK
| | | | - Hans-Christoph Diener
- Institute for Medical Informatics, Biometry and Epidemiology, Medical Faculty of the University Duisburg-Essen, Duisburg-Essen, Germany
| | | |
Collapse
|
31
|
Okue S, Yaguchi M, Miura A, Ozaki-Masuzawa Y, Hosono T, Seki T. The garlic-derived organosulfur compound diallyl trisulphide suppresses tissue factor function. Food Funct 2022; 13:1246-1255. [PMID: 35022635 DOI: 10.1039/d1fo02206g] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tissue factor (TF) is a critical initiator of extrinsic coagulation that sometimes causes thromboembolism. Diallyl trisulphide (DATS) is a secondary metabolite of allicin generated in crushed garlic, with various pharmacological effects. This study aimed to clarify the effect of DATS on the extrinsic coagulation elicited by TF and arteriosclerosis. TF activity was measured using a clotting assay in TF-expressing HL60 cells. DATS inhibited TF activity in a dose-dependent manner. TF expression in TNF-α-stimulated human umbilical vein endothelial cells was examined using real-time PCR and western blotting. DATS inhibited TF mRNA and protein expression induced by TNF-α via inhibition of JNK signalling. The effect of DATS on arteriosclerosis was also examined in apolipoprotein E-deficient mice. DATS administration in these mice tended to decrease atherosclerotic lesion size. These results strongly suggest that DATS prevents thromboembolism triggered by atherosclerosis via the inhibition of plaque formation and TF function.
Collapse
Affiliation(s)
- Sachiko Okue
- Department of Applied Life Sciences, Nihon University Graduate School of Bioresource Sciences, Kanagawa, Japan.
| | - Manami Yaguchi
- Department of Applied Life Sciences, Nihon University Graduate School of Bioresource Sciences, Kanagawa, Japan.
| | - Atsushi Miura
- Department of Applied Life Sciences, Nihon University Graduate School of Bioresource Sciences, Kanagawa, Japan.
| | - Yori Ozaki-Masuzawa
- Department of Chemistry and Life Science, Collage of Bioresource Sciences, Nihon University, Kanagawa, Japan
| | - Takashi Hosono
- Department of Applied Life Sciences, Nihon University Graduate School of Bioresource Sciences, Kanagawa, Japan. .,Department of Chemistry and Life Science, Collage of Bioresource Sciences, Nihon University, Kanagawa, Japan
| | - Taiichiro Seki
- Department of Applied Life Sciences, Nihon University Graduate School of Bioresource Sciences, Kanagawa, Japan. .,Department of Chemistry and Life Science, Collage of Bioresource Sciences, Nihon University, Kanagawa, Japan
| |
Collapse
|
32
|
Kosiński K, Malinowski D, Safranow K, Dziedziejko V, Pawlik A. PECAM1, COL4A2, PHACTR1, and LMOD1 Gene Polymorphisms in Patients with Unstable Angina. J Clin Med 2022; 11:jcm11020373. [PMID: 35054067 PMCID: PMC8778316 DOI: 10.3390/jcm11020373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/07/2022] [Accepted: 01/11/2022] [Indexed: 11/16/2022] Open
Abstract
Coronary artery disease (CAD) is a syndrome resulting from myocardial ischaemia of heterogeneous pathomechanism. Environmental and genetic factors contribute to its development. Atherosclerotic plaques that significantly narrow the lumen of coronary arteries cause symptoms of myocardial ischaemia. Acute coronary incidents are most often associated with plaque rupture or erosion accompanied by local activation of the coagulation system with thrombus formation. Plaque formation and stability are influenced by endothelial function and vascular smooth muscle cell function. In this study, we investigated the association between polymorphisms in genes affecting endothelial and vascular smooth muscle cell (VSMC) function and the occurrence of unstable angina pectoris. The aim of this study was to evaluate the association between the PECAM1 (rs1867624), COL4A2 (rs4773144), PHACTR1 (rs9349379) and LMOD1 (rs2820315) gene polymorphisms and the risk of unstable angina. The study included 232 patients with unstable angina diagnosed on the basis of clinical symptoms and coronary angiography and 144 healthy subjects with no significant coronary lumen stenosis at coronary angiography. There were no statistically significant differences in the distribution of COL4A2 rs4773144 and PECAM1 rs1867624 gene polymorphisms between patients with unstable angina and control subjects. In patients with unstable angina, there was an increased frequency of PHACTR1 rs9349379 G allele carriers (GG and AG genotypes) (GG+AG vs. AA, OR 1.71; 95% CI 1.10-2.66, p = 0.017) and carriers of the LMOD1 rs2820315 T allele (TT and CT genotypes) (TT+CT vs. CC, OR 1.65; 95% CI 1.09-2.51, p = 0.019) compared to the control group. The association between these alleles and unstable angina was confirmed by multivariate logistic regression analysis, in which the number of G (PHACTR1 rs9349379) and T (LMOD1 rs2820315) alleles was an independent risk factor for unstable angina. The results suggest an association between PHACTR1 rs9349379 and LMOD1 rs2820315 polymorphisms and the risk of unstable angina.
Collapse
Affiliation(s)
- Krzysztof Kosiński
- Department of Cardiology, Hospital in Szczecin, Arkonska 4, 71-455 Szczecin, Poland;
| | - Damian Malinowski
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, 70-111 Szczecin, Poland;
| | - Krzysztof Safranow
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.S.); (V.D.)
| | - Violetta Dziedziejko
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.S.); (V.D.)
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
- Correspondence:
| |
Collapse
|
33
|
Lim XC, Yatim SMJM, Chong SY, Wang X, Tan SH, Yang X, Chan SP, Richards AM, Charles CJ, Chan MY, Wang JW. Plasma tissue factor coagulation activity in post-acute myocardial infarction patients. Front Endocrinol (Lausanne) 2022; 13:1008329. [PMID: 36213278 PMCID: PMC9540383 DOI: 10.3389/fendo.2022.1008329] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/08/2022] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION Coagulation is involved in fibroproliferative responses following acute myocardial infarction (AMI). Left ventricular (LV) remodeling following AMI is closely associated with progression to heart failure. This study aims to assess the association between plasma tissue factor activity and LV remodeling in post-AMI patients. METHODS We studied 228 patients with AMI and 57 healthy subjects. Patients with AMI were categorized into two age- and sex-matched groups: patients with adverse LV remodeling or reverse LV remodeling, defined by an increase or decrease, respectively, in LV end systolic volume by ≥15% over 6 months. TF activity was measured in plasma collected at baseline (within 72 hours of revascularization), 1 month and 6 months post-AMI. Multiple level longitudinal data analysis with structural equation (ML-SEM) model was used to assess the impact of various clinical variables on TF activity in post-AMI. RESULTS Plasma TF activity in post-AMI patients at baseline (29.05 ± 10.75 pM) was similar to that in healthy subjects but fell at 1 month (21.78 ± 8.23, p<0.001) with partial recovery by 6 months (25.84 ± 8.80, p<0.001) after AMI. Plasma TF activity at 6 month post-AMI was better restored in patients with reverse LV remodeling than those with adverse LV remodeling (27.35 ± 7.14 vs 24.34 ± 9.99; p=0.009) independent of gender, age and relevant cardiovascular risk factors. CONCLUSIONS Plasma TF activity decreased after AMI but was better restored at 6 months in patients with reverse LV remodeling. The clinical significance of changes in post-AMI plasma TF activity needs further investigation.
Collapse
Affiliation(s)
- Xiong Chang Lim
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Siti Maryam J. M. Yatim
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cardiovascular Research Institute, National University Heart Centre Singapore, Singapore, Singapore
| | - Suet Yen Chong
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cardiovascular Research Institute, National University Heart Centre Singapore, Singapore, Singapore
| | - Xiaoyuan Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cardiovascular Research Institute, National University Heart Centre Singapore, Singapore, Singapore
| | - Sock Hwee Tan
- Cardiovascular Research Institute, National University Heart Centre Singapore, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xiaoxun Yang
- Cardiovascular Research Institute, National University Heart Centre Singapore, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Siew Pang Chan
- Cardiovascular Research Institute, National University Heart Centre Singapore, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - A Mark Richards
- Cardiovascular Research Institute, National University Heart Centre Singapore, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chris J. Charles
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cardiovascular Research Institute, National University Heart Centre Singapore, Singapore, Singapore
| | - Mark Y. Chan
- Cardiovascular Research Institute, National University Heart Centre Singapore, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- *Correspondence: Jiong-Wei Wang, ; Mark Y. Chan,
| | - Jiong-Wei Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cardiovascular Research Institute, National University Heart Centre Singapore, Singapore, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- *Correspondence: Jiong-Wei Wang, ; Mark Y. Chan,
| |
Collapse
|
34
|
MOLECULAR MECHANISMS OF INTRAVASCULAR INHIBITION AND STIMULATION OF EXTRAVASCULAR THROMBOSIS. BIOTECHNOLOGIA ACTA 2021. [DOI: 10.15407/biotech14.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The hemostasis system is designed to maintain a balance between the processes of blood clotting, anticoagulation, as well as fibrinolysis, to ensure constant effective blood circulation in the body and rapid cessation of bleeding in the event of their occurrence. The procoagulant potential of the hemostasis system is based on molecular mechanisms that lead to the formation of fibrin in the bloodstream, which is the framework of the thrombus, and to the aggregation of platelets — the basis of the thrombus body. The anticoagulant potential of blood plasma is provided by mechanisms aimed at inhibiting blood coagulation processes. Thorough study and understanding of these mechanisms will open up numerous treatments for pathologies associated with both intravascular thrombosis and bleeding of various origins. The purpose of this review is to analyze ways to prevent intravascular thrombosis and stimulate extravascular thrombosis. The review describes and analyzes available and promising means of thrombosis prevention, in particular, direct and indirect anticoagulants and antiplatelets, as well as methods of effective stimulation of thrombosis, which is necessary in case of vascular damage. The result of this analysis is to determine the nodal points of the protein network of the hemostasis system, the action of which by specific molecular effectors will control the process of thrombosis.
Collapse
|
35
|
Deter HC, Orth-Gomér K, Rauch-Kröhnert U, Albus C, Ladwig KH, Söllner W, de Zwaan M, Grün AS, Ronel J, Hellmich M, Herrmann-Lingen C, Weber C. Depression, anxiety, and vital exhaustion are associated with pro-coagulant markers in depressed patients with coronary artery disease - A cross sectional and prospective secondary analysis of the SPIRR-CAD trial. J Psychosom Res 2021; 151:110659. [PMID: 34763203 DOI: 10.1016/j.jpsychores.2021.110659] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 10/22/2021] [Accepted: 10/24/2021] [Indexed: 01/08/2023]
Abstract
INTRODUCTION A hyper-coagulant state is a biological mechanism that triggers cardiac events in patients with coronary artery disease (CAD). Depressive symptoms and anxiety predict an unfavourable course of CAD. The SPIRR-CAD-RCT examined the effects of a psychological intervention and provided the opportunity to explore cross-sectional associations between indices of psychological strain and coagulation parameters, as well as prospective changes in depression scores and coagulation parameters. METHODS In this secondary analysis, we investigated 253 CAD patients (194 male; age m 58.9, SD 8.3 yrs.) with mild to moderate depression (≥8 on the HADS-D) at baseline and at follow-up 18 months later: TF, fibrinogen, D-dimer, VWF, FVII and PAI-1 and the course of depression (HAM-D), vital exhaustion (VE) and anxiety scores (HADS-A) were examined by ANOVA in the total and younger age groups (≤ 60). RESULTS HAM-D at baseline was correlated with TF (corr. R2 = 0.27; F = 9.31, p = 0.001). HADS anxiety was associated with fibrinogen (corr. R2.20; F = 7.27, p = 0.001). There was no detectable therapeutic effect on coagulation. Fibrinogen and VWF decreased within 18 months (time effect; p = 0.02; p = 0.04), as did HADS-D in both treatment groups (p < 0.001). Fibrinogen decreased more in patients ≤60 years with high VE compared to low VE (interaction time x group, p = 0.01). CONCLUSIONS This is the first study to show an association between TF and depression. Coagulation parameters as potential mediators of CAD progression correlated cross-sectionally with depression and anxiety and prospectively with VE. Further studies should replicate these correlations in depressed and non-depressed CAD patients. ISRCTN 76240576; clinicaltrials.gov.
Collapse
Affiliation(s)
- Hans-Christian Deter
- Medical Clinic, Psychosomatics, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, German Center for Cardiovascular Research, Partner Site Berlin, Germany.
| | | | - Ursula Rauch-Kröhnert
- Medical Clinic, Cardiology and Pulmonology, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Germany
| | - Christian Albus
- Department of Psychosomatics and Psychotherapy, University of Cologne, Germany
| | - Karl-Heinz Ladwig
- Department of Psychosomatic Medicine and Psychotherapy, Klinikum Rechts der Isar, Technische Universität München (TUM), German Center for Cardiovascular Research, Partner Site Munich, Germany
| | - Wolfgang Söllner
- Department of Psychosomatic Medicine and Psychotherapy, Paracelsus Medical University, Nuremberg General Hospital, Germany
| | - Martina de Zwaan
- Department of Psychosomatic Medicine and Psychotherapy, Hannover Medical School, Germany
| | - Anna-Sophia Grün
- Medical Clinic, Psychosomatics, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, German Center for Cardiovascular Research, Partner Site Berlin, Germany
| | - Joram Ronel
- Klinik Barmelweid, Switzerland; Department of Psychosomatic Medicine and Psychotherapy, Klinikum Rechts der Isar, Technische Universität München, Germany
| | - Martin Hellmich
- Clinical Trials Center Cologne, Institute for Medical Statistics, Informatic und Epidemiology (IMSIE), University of Cologne, Germany
| | - Christoph Herrmann-Lingen
- Dept. of Psychosomatic Medicine and Psychotherapy, University of Göttingen Medical Center, German Center for Cardiovascular Research, Partner Site Göttingen, Germany
| | - Cora Weber
- Medical Clinic, Psychosomatics, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, German Center for Cardiovascular Research, Partner Site Berlin, Germany
| | | |
Collapse
|
36
|
Ma Z, Yang KY, Huang Y, Lui KO. Endothelial contribution to COVID-19: an update on mechanisms and therapeutic implications. J Mol Cell Cardiol 2021; 164:69-82. [PMID: 34838588 PMCID: PMC8610843 DOI: 10.1016/j.yjmcc.2021.11.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 12/15/2022]
Abstract
The global propagation of SARS-CoV-2 leads to an unprecedented public health emergency. Despite that the lungs are the primary organ targeted by COVID-19, systemic endothelial inflammation and dysfunction is observed particularly in patients with severe COVID-19, manifested by elevated endothelial injury markers, endotheliitis, and coagulopathy. Here, we review the clinical characteristics of COVID-19 associated endothelial dysfunction; and the likely pathological mechanisms underlying the disease including direct cell entry or indirect immune overreactions after SARS-CoV-2 infection. In addition, we discuss potential biomarkers that might indicate the disease severity, particularly related to the abnormal development of thrombosis that is a fatal vascular complication of severe COVID-19. Furthermore, we summarize clinical trials targeting the direct and indirect pathological pathways after SARS-CoV-2 infection to prevent or inhibit the virus induced endothelial disorders.
Collapse
Affiliation(s)
- Zhangjing Ma
- Department of Chemical Pathology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Kevin Y Yang
- Department of Chemical Pathology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Kathy O Lui
- Department of Chemical Pathology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China; Li Ka Shing Institute of Health Science, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China; Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.
| |
Collapse
|
37
|
Rivera Rivas JJ, Czuprynski CJ. Histophilus somni stimulates bovine monocyte-derived macrophages to release microparticles that increase fibrin clot formation in vitro. Vet Microbiol 2021; 264:109280. [PMID: 34808430 DOI: 10.1016/j.vetmic.2021.109280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 10/14/2021] [Accepted: 11/08/2021] [Indexed: 11/28/2022]
Abstract
Histophilus somni is a Gram-negative coccobacillus that causes diffuse vasculitis and intravascular thrombosis that can lead to multiple organ failure in cattle. Macrophages are important cellular mediators of fibrin deposition and removal at sites of inflammation. It has become evident that macrophages and other cells release microparticles (MPs) that have an array of biological activities, including pro-coagulant activity. We sought to determine whether monocyte-derived macrophages exposed to H. somni in vitro release MPs that activate the clotting cascade in a manner that could lead to thrombus formation. Bovine monocyte-derived macrophages were incubated with H. somni (at a 10:1 ratio) in RPMI with 10% heat inactivated fetal bovine serum for 6 h at 37 °C with 5 % CO2. Membrane-shed MPs were isolated from the conditioned media, washed twice with Ca2+ and Mg2+ free HBSS, and pro-coagulant activity assessed by a one-step plasma clotting assay. We observed greater pro-coagulant activity for MPs from H. somni stimulated macrophages than from unstimulated controls. Microparticle pro-coagulant activity was inhibited by addition of an anti-tissue factor antibody. We also observed co-localization of fluorescein-labeled H. somni cells and annexin V staining as evaluated by confocal microscopy. These results demonstrate that exposure to H. somni cells causes bovine monocyte-derived macrophages to release MPs that contain tissue factor, the first such report for bovine macrophages. We infer that if similar events occur in vivo they could amplify thrombus formation in bovine histophilosis.
Collapse
Affiliation(s)
- José J Rivera Rivas
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI, 53706, USA.
| | - Charles J Czuprynski
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI, 53706, USA
| |
Collapse
|
38
|
Rivaroxaban attenuates cardiac hypertrophy by inhibiting protease-activated receptor-2 signaling in renin-overexpressing hypertensive mice. Hypertens Res 2021; 44:1261-1273. [PMID: 34285375 DOI: 10.1038/s41440-021-00700-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 03/07/2021] [Accepted: 03/28/2021] [Indexed: 02/07/2023]
Abstract
Rivaroxaban (Riv), a direct factor Xa (FXa) inhibitor, exerts anti-inflammatory effects in addition to anticoagulation. However, its role in cardiovascular remodeling is largely unknown. We tested the hypothesis that Riv attenuates the progression of cardiac hypertrophy and fibrosis induced by continuous activation of the renin-angiotensin system (RAS) in renin-overexpressing hypertensive transgenic (Ren-Tg) mice. We treated 12-week-old male Ren-Tg and wild-type (WT) mice with a diet containing Riv (12 mg/kg/day) or a regular diet for 4 weeks. After this, FXa in plasma significantly increased in Ren-Tg mice compared with WT mice, and Riv inhibited this increase. Left ventricular wall thickness (LVWT) and the area of cardiac fibrosis evaluated by Masson's trichrome staining were greater in Ren-Tg mice than in WT mice, and Riv decreased them. Cardiac expression levels of the protease-activated receptor (PAR)-2, tumor necrosis factor-α, transforming growth factor (TGF)-β1, and collagen type 3 α1 (COL3A1) genes were all greater in Ren-Tg mice than in WT mice, and Riv attenuated these increases. To investigate the possible involvement of PAR-2, we treated Ren-Tg mice with a continuous subcutaneous infusion of 10 μg/kg/day of the PAR-2 antagonist FSLLRY for 4 weeks. FSLLRY significantly decreased LVWT and cardiac expression of PAR-2, TGF-β1, and COL3A1. In isolated cardiac fibroblasts (CFs), Riv or FSLLRY pretreatment inhibited the FXa-induced increase in the phosphorylation of extracellular signal-regulated kinases. In addition, Riv or FSLLRY inhibited FXa-stimulated wound closure in CFs. Riv exerts a protective effect against cardiac hypertrophy and fibrosis development induced by continuous activation of the RAS, partly by inhibiting PAR-2.
Collapse
|
39
|
Bobescu E, Marceanu LG, Covaciu A, Vladau LA. Thrombosis, an important piece in the COVID-19 puzzle: From pathophysiology to therapy. Anatol J Cardiol 2021; 25:601-608. [PMID: 34498590 DOI: 10.5152/anatoljcardiol.2021.475] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
A lot of data about coronavirus disease 2019 (COVID-19) have been already published; however, these still form only a part of the pandemic puzzle. Once we have all the pieces of the puzzle, we will be able to successfully treat this disease with its multiple challenges. COVID-19 has a high thrombogenic potential. In this study, we aimed to review published data about COVID-19 associated thrombosis from pathophysiology to treatment and the role in patient evolution. We searched for articles and studies published online through MEDLINE/PubMed database, Google Scholar, ScienceDirect, Wiley Online Library, and Nature Public Health Emergency Collection. We found numerous articles regarding COVID-19 infection but selected only those focused on thrombosis. D-dimers have a predictive value in identifying thrombosis and a high level correlates with the severity of the infection and death. Most patients who were on chronic anticoagulant therapy before contracting the virus had a better prognosis. Heparin has other favorable effects such as a direct antiviral and anti-inflammatory effect in addition to its anticoagulant effect. COVID-19 infections are frequently complicated by thrombotic pathology. High plasma level of D-dimers is a predictive factor for severe prognosis, and the recommended anticoagulant, associated with low mortality, is heparin followed by a direct oral anticoagulant. Randomized studies in large groups of patients and therapeutic guidelines are still needed on this subject.
Collapse
Affiliation(s)
- Elena Bobescu
- Department of Medical and Surgical Specialties, Faculty of Medicine, Transilvania University of Brasov; Brasov- Romania;Department of Cardiology, Clinical County Emergency Hospital Brasov; Brasov-Romania
| | - Luigi Geo Marceanu
- Department of Medical and Surgical Specialties, Faculty of Medicine, Transilvania University of Brasov; Brasov- Romania
| | - Alexandru Covaciu
- Department of Medical and Surgical Specialties, Faculty of Medicine, Transilvania University of Brasov; Brasov- Romania;Department of Cardiology, Clinical County Emergency Hospital Brasov; Brasov-Romania
| | - Larisa Alexandra Vladau
- Department of Medical and Surgical Specialties, Faculty of Medicine, Transilvania University of Brasov; Brasov- Romania;Department of Cardiology, Clinical County Emergency Hospital Brasov; Brasov-Romania
| |
Collapse
|
40
|
Nakano T, Takahashi T, Yamamoto C, Kaji T, Fujiwara Y. Arsenite induces tissue factor synthesis through Nrf2 activation in cultured human aortic smooth muscle cells. J Toxicol Sci 2021; 46:187-192. [PMID: 33814512 DOI: 10.2131/jts.46.187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Tissue factor (TF) is the initiator of the coagulation cascade, constitutively expressed in subendothelial cells such as vascular smooth muscle cells and initiating rapid coagulation when the vascular vessel is damaged. TF has been shown to be involved in the development and progression of atherosclerosis. Arsenic, an environmental pollutant, is related to the progression of atherosclerosis, although the pathogenic mechanisms are not fully elucidated. In the present study, we investigated the effect of arsenite on the expression of TF in human aortic smooth muscle cells (HASMCs) and the underlying molecular mechanisms. We found that (1) arsenite stimulated TF synthesis and activated the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway in HASMCs, (2) sulforaphane, an Nrf2 activator, also stimulated TF synthesis in HASMCs, and (3) arsenite-induced upregulation of TF synthesis was prevented by Nrf2 knockdown in HASMCs. These results suggest that arsenite promotes TF synthesis by activating the Nrf2 pathway in HASMCs and that the induction of TF expression by arsenite may be related to the progression of atherosclerosis.
Collapse
Affiliation(s)
- Tsuyoshi Nakano
- Department of Environmental Health, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Tsutomu Takahashi
- Department of Environmental Health, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Chika Yamamoto
- Department of Environmental Health, Faculty of Pharmaceutical Sciences, Toho University
| | - Toshiyuki Kaji
- Department of Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science
| | - Yasuyuki Fujiwara
- Department of Environmental Health, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| |
Collapse
|
41
|
Mackiewicz-Milewska M, Kroszczynski A, Cisowska-Adamiak M, Pyskir J, Rosc D, Hagner W. Hemostatic parameters in patients with spinal cord injury in subacute and chronic phase of the rehabilitation. J Spinal Cord Med 2021; 44:782-788. [PMID: 32011973 PMCID: PMC8477962 DOI: 10.1080/10790268.2019.1708600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Objective: The goal of this study was to measure hemostatic markers after SCI.Design: Assesing changes in coagulation and fibrynilitic system in SCI patients in different time post injury to Cross-sectional study.Setting: Rehabilitation Department of the Bydgoszcz University Hospital, Poland from 2011 to 2017.Participants: SCI patient during acute and chronic rehabilitation (N = 88).Outcome Measures: Assesing following parameters: platelet counts and levels of D-dimer, antithrombin III (ATIII), tissue factor (TF), tissue factor pathway inhibitor (TFPI) and the inflammatory marker, C-reactive protein (CRP).Interventions: Eighty-eight SCI patients were divided into three groups based on the time elapsed from injury: group I (three weeks to three months), group II (three to twelve months) and group III (more than twelve months). All patients underwent ultrasonography (US) to detect acute or chronic recanalized deep vein thrombosis (DVT). Platelet counts and levels of D-dimer, ATIII, TF, TFPI and CRP were assessed. TF and TFPI levels were measured in the control group of forty healthy individuals without SCI, the rest of the parameters were compared to laboratory norms.Results: D-dimer levels were significantly higher in group I compared to group II (P = .0002) and group III (P < .001). Group II had higher D-dimer levels than group III (P = .032). TFPI levels were higher in group II compared with group III (P = .0041) and control group (P = .000033). TF was significantly higher in all the SCI groups compared with the control group (P < .001).Conclusions: D-dimer and TF levels were still elevated twelve months after SCI. TF levels were also elevated over 12 months after inury. The results may indicate that sub-acute and even chronic SCI patients have disturbed coagulation and fibrynolitic system.
Collapse
Affiliation(s)
- Magdalena Mackiewicz-Milewska
- Department of Rehabilitation Collegium Medicum in Bydgoszcz, Faculty of Health Science, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland,Correspondence to: Magdalena Mackiewicz-Milewska, University Hospital no.1 in Bydgoszcz, Skłodowskiej-Curie 9 Street, Bydgoszcz85–091, Poland; Ph: +48 52 5854674.
| | | | - Małgorzata Cisowska-Adamiak
- Department of Rehabilitation Collegium Medicum in Bydgoszcz, Faculty of Health Science, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| | - Jerzy Pyskir
- Department of Biophysics Collegium Medicum in Bydgoszcz, Faculty of Pharmacy, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| | - Danuta Rosc
- Department of Pathophysiology Collegium Medicum in Bydgoszcz, Faculty of Pharmacy Nicolaus, Copernicus University, Bydgoszcz, Poland
| | - Wojciech Hagner
- Department of Rehabilitation Collegium Medicum in Bydgoszcz, Faculty of Health Science, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| |
Collapse
|
42
|
Migdalski A, Jawien A. New insight into biology, molecular diagnostics and treatment options of unstable carotid atherosclerotic plaque: a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1207. [PMID: 34430648 PMCID: PMC8350668 DOI: 10.21037/atm-20-7197] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 05/24/2021] [Indexed: 12/23/2022]
Abstract
Indications for intervention in hemodynamically relevant carotid artery stenosis (carotid endarterectomy or stenting) are primarily based on a degree of stenosis and symptomatology. To date the plaque vulnerability is rarely taken into account in clinical decision making although development of molecular imaging allows a better understanding of plaque biology and provides new techniques detecting potentially vulnerable plaque at risk. A significant number of reports describing the mechanisms of unstable plaque formation suggest that it is a multifactorial process. Inflammation, lipid accumulation, apoptosis, proteolysis, the thrombotic process and angiogenesis are among the main factors of carotid plaque destabilization. Although inflammation is a key process in development of plaque vulnerability, the hemostasis and neoangiogenesis should be regarded as equally important. Only a small group of asymptomatic patients may benefit from the invasive treatment and it remains a challenge to determine whether initially asymptomatic carotid plaque become unstable or vulnerable. Currently, the main task of research on atherosclerotic lesion imaging is focused on functional state of the plaque. The presence of one or more features such as stenosis progression, large plaque area, large juxta-luminal black area, plaque echolucency, intra-plaque hemorrhage, impaired cerebral vascular reserve and spontaneous embolization may indicate patients at higher risk for stroke suitable for revascularization. Treatment of carotid stenosis as one of the manifestations of generalized atherosclerosis requires a broad approach. Nowadays pharmacological treatment options for the atherosclerotic process are largely aimed at stimulating the plaque stabilization, but in symptomatic patients and selected asymptomatic patients, carotid plaque should be removed as a potential source of embolism.
Collapse
Affiliation(s)
- Arkadiusz Migdalski
- Department of Vascular Surgery and Angiology, L. Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Arkadiusz Jawien
- Department of Vascular Surgery and Angiology, L. Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| |
Collapse
|
43
|
Witkowski M, Witkowski M, Friebel J, Buffa JA, Li XS, Wang Z, Sangwan N, Li L, DiDonato JA, Tizian C, Haghikia A, Kirchhofer D, Mach F, Räber L, Matter CM, Tang WHW, Landmesser U, Lüscher TF, Rauch U, Hazen SL. Vascular endothelial tissue factor contributes to trimethylamine N-oxide-enhanced arterial thrombosis. Cardiovasc Res 2021; 118:2367-2384. [PMID: 34352109 PMCID: PMC9890461 DOI: 10.1093/cvr/cvab263] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 08/02/2021] [Indexed: 02/04/2023] Open
Abstract
AIMS Gut microbiota and their generated metabolites impact the host vascular phenotype. The metaorganismal metabolite trimethylamine N-oxide (TMAO) is both associated with adverse clinical thromboembolic events, and enhances platelet responsiveness in subjects. The impact of TMAO on vascular Tissue Factor (TF) in vivo is unknown. Here, we explore whether TMAO-enhanced thrombosis potential extends beyond TMAO effects on platelets, and is linked to TF. We also further explore the links between gut microbiota and vascular endothelial TF expression in vivo. METHODS AND RESULTS In initial exploratory clinical studies, we observed that among sequential stable subjects (n = 2989) on anti-platelet therapy undergoing elective diagnostic cardiovascular evaluation at a single-site referral centre, TMAO levels were associated with an increased incident (3 years) risk for major adverse cardiovascular events (MACE) (myocardial infarction, stroke, or death) [4th quartile (Q4) vs. Q1 adjusted hazard ratio (HR) 95% confidence interval (95% CI), 1.73 (1.25-2.38)]. Similar results were observed within subjects on aspirin mono-therapy during follow-up [adjusted HR (95% CI) 1.75 (1.25-2.44), n = 2793]. Leveraging access to a second higher risk cohort with previously reported TMAO data and monitoring of anti-platelet medication use, we also observed a strong association between TMAO and incident (1 year) MACE risk in the multi-site Swiss Acute Coronary Syndromes Cohort, focusing on the subset (n = 1469) on chronic dual anti-platelet therapy during follow-up [adjusted HR (95% CI) 1.70 (1.08-2.69)]. These collective clinical data suggest that the thrombosis-associated effects of TMAO may be mediated by cells/factors that are not inhibited by anti-platelet therapy. To test this, we first observed in human microvascular endothelial cells that TMAO dose-dependently induced expression of TF and vascular cell adhesion molecule (VCAM)1. In mouse studies, we observed that TMAO-enhanced aortic TF and VCAM1 mRNA and protein expression, which upon immunolocalization studies, was shown to co-localize with vascular endothelial cells. Finally, in arterial injury mouse models, TMAO-dependent enhancement of in vivo TF expression and thrombogenicity were abrogated by either a TF-inhibitory antibody or a mechanism-based microbial choline TMA-lyase inhibitor (fluoromethylcholine). CONCLUSION Endothelial TF contributes to TMAO-related arterial thrombosis potential, and can be specifically blocked by targeted non-lethal inhibition of gut microbial choline TMA-lyase.
Collapse
Affiliation(s)
- Marco Witkowski
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, 9500 Euclid Ave, Cleveland, OH 44195, USA,Department of Cardiology, Charité Centrum 11, Charité–Universitätsmedizin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Mario Witkowski
- Department of Microbiology, Infectious Diseases and Immunology, Laboratory of Innate Immunity, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Julian Friebel
- Department of Cardiology, Charité Centrum 11, Charité–Universitätsmedizin, Hindenburgdamm 30, 12203, Berlin, Germany,Berlin Institute of Health, Anna-Louisa-Karsch-Straße 2, 10178, Berlin, Germany
| | - Jennifer A Buffa
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Xinmin S Li
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Zeneng Wang
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Naseer Sangwan
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Lin Li
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Joseph A DiDonato
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Caroline Tizian
- Department of Microbiology, Infectious Diseases and Immunology, Laboratory of Innate Immunity, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Arash Haghikia
- Department of Cardiology, Charité Centrum 11, Charité–Universitätsmedizin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Daniel Kirchhofer
- Department of Early Discovery Biochemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - François Mach
- Department of Cardiology, University Hospital Geneva, Rue Gabrielle-Perret-Gentil 4 1205, Geneva, Switzerland
| | - Lorenz Räber
- Department of Cardiology, Inselspital Bern, Freiburgstrasse 18 CH-3010, Bern, Switzerland
| | - Christian M Matter
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, CH-8952 Schlieren, Switzerland,Department of Cardiology, University Heart Center, University Hospital Zurich, Raemistrasse 100 8091, Zurich, Switzerland
| | - W H Wilson Tang
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, 9500 Euclid Ave, Cleveland, OH 44195, USA,Department of Cardiovascular Medicine, Heart, Vascular & Thoracic Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH, USA
| | - Ulf Landmesser
- Department of Cardiology, Charité Centrum 11, Charité–Universitätsmedizin, Hindenburgdamm 30, 12203, Berlin, Germany,Berlin Institute of Health, Anna-Louisa-Karsch-Straße 2, 10178, Berlin, Germany
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, CH-8952 Schlieren, Switzerland,Department of Cardiology, Royal Brompton and Harefield Hospitals, Imperial College, Sydney St, London SW3 6NP, UK
| | - Ursula Rauch
- Corresponding author. Tel: +1 216 445 9763; fax: +1 216 444 9404, E-mail: (S.L.H.); Tel: +49 30 8445 2362; fax: +49 30 8445 4648, E-mail: (U.R.)
| | - Stanley L Hazen
- Corresponding author. Tel: +1 216 445 9763; fax: +1 216 444 9404, E-mail: (S.L.H.); Tel: +49 30 8445 2362; fax: +49 30 8445 4648, E-mail: (U.R.)
| |
Collapse
|
44
|
Kraler S, Libby P, Evans PC, Akhmedov A, Schmiady MO, Reinehr M, Camici GG, Lüscher TF. Resilience of the Internal Mammary Artery to Atherogenesis: Shifting From Risk to Resistance to Address Unmet Needs. Arterioscler Thromb Vasc Biol 2021; 41:2237-2251. [PMID: 34107731 PMCID: PMC8299999 DOI: 10.1161/atvbaha.121.316256] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Fueled by the global surge in aging, atherosclerotic cardiovascular disease reached pandemic dimensions putting affected individuals at enhanced risk of myocardial infarction, stroke, and premature death. Atherosclerosis is a systemic disease driven by a wide spectrum of factors, including cholesterol, pressure, and disturbed flow. Although all arterial beds encounter a similar atherogenic milieu, the development of atheromatous lesions occurs discontinuously across the vascular system. Indeed, the internal mammary artery possesses unique biological properties that confer protection to intimal growth and atherosclerotic plaque formation, thus making it a conduit of choice for coronary artery bypass grafting. Its endothelium abundantly expresses nitric oxide synthase and shows accentuated nitric oxide release, while its vascular smooth muscle cells exhibit reduced tissue factor expression, high tPA (tissue-type plasminogen activator) production and blunted migration and proliferation, which may collectively mitigate intimal thickening and ultimately the evolution of atheromatous plaques. We aim here to provide insights into the anatomy, physiology, cellular, and molecular aspects of the internal mammary artery thereby elucidating its remarkable resistance to atherogenesis. We propose a change in perspective from risk to resilience to decipher mechanisms of atheroresistance and eventually identification of novel therapeutic targets presently not addressed by currently available remedies.
Collapse
Affiliation(s)
- Simon Kraler
- Center for Molecular Cardiology, University of Zürich, 8952 Schlieren, Switzerland
| | - Peter Libby
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA
| | - Paul C. Evans
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Alexander Akhmedov
- Center for Molecular Cardiology, University of Zürich, 8952 Schlieren, Switzerland
| | - Martin O. Schmiady
- Clinic for Cardiac Surgery, University Heart Centre, University Hospital Zurich, Zurich, Switzerland
| | - Michael Reinehr
- Institute of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Giovanni G. Camici
- Center for Molecular Cardiology, University of Zürich, 8952 Schlieren, Switzerland
- University Heart Center, Department of Cardiology, University Hospital, Zurich, Switzerland
- Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Thomas F. Lüscher
- Center for Molecular Cardiology, University of Zürich, 8952 Schlieren, Switzerland
- Royal Brompton and Harefield Hospitals and Imperial College, London, United Kingdom
| |
Collapse
|
45
|
Das K, Keshava S, Ansari SA, Kondreddy V, Esmon CT, Griffin JH, Pendurthi UR, Rao LVM. Factor VIIa induces extracellular vesicles from the endothelium: a potential mechanism for its hemostatic effect. Blood 2021; 137:3428-3442. [PMID: 33534910 PMCID: PMC8212509 DOI: 10.1182/blood.2020008417] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 01/11/2021] [Indexed: 12/14/2022] Open
Abstract
Recombinant factor FVIIa (rFVIIa) is used as a hemostatic agent to treat bleeding disorders in hemophilia patients with inhibitors and other groups of patients. Our recent studies showed that FVIIa binds endothelial cell protein C receptor (EPCR) and induces protease-activated receptor 1 (PAR1)-mediated biased signaling. The importance of FVIIa-EPCR-PAR1-mediated signaling in hemostasis is unknown. In the present study, we show that FVIIa induces the release of extracellular vesicles (EVs) from endothelial cells both in vitro and in vivo. Silencing of EPCR or PAR1 in endothelial cells blocked the FVIIa-induced generation of EVs. Consistent with these data, FVIIa treatment enhanced the release of EVs from murine brain endothelial cells isolated from wild-type (WT), EPCR-overexpressing, and PAR1-R46Q-mutant mice, but not EPCR-deficient or PAR1-R41Q-mutant mice. In vivo studies revealed that administration of FVIIa to WT, EPCR-overexpressing, and PAR1-R46Q-mutant mice, but not EPCR-deficient or PAR1-R41Q-mutant mice, increased the number of circulating EVs. EVs released in response to FVIIa treatment exhibit enhanced procoagulant activity. Infusion of FVIIa-generated EVs and not control EVs to platelet-depleted mice increased thrombin generation at the site of injury and reduced blood loss. Administration of FVIIa-generated EVs or generation of EVs endogenously by administering FVIIa augmented the hemostatic effect of FVIIa. Overall, our data reveal that FVIIa treatment, through FVIIa-EPCR-PAR1 signaling, releases EVs from the endothelium into the circulation, and these EVs contribute to the hemostatic effect of FVIIa.
Collapse
Affiliation(s)
- Kaushik Das
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX
| | - Shiva Keshava
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX
| | - Shabbir A Ansari
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX
| | - Vijay Kondreddy
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX
| | - Charles T Esmon
- Coagulation Biology Laboratory, Oklahoma Medical Research Foundation, Oklahoma City, OK; and
| | - John H Griffin
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA
| | - Usha R Pendurthi
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX
| | - L Vijaya Mohan Rao
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX
| |
Collapse
|
46
|
Deng H, Tang TX, Chen D, Tang LS, Yang XP, Tang ZH. Endothelial Dysfunction and SARS-CoV-2 Infection: Association and Therapeutic Strategies. PATHOGENS (BASEL, SWITZERLAND) 2021; 10:pathogens10050582. [PMID: 34064553 PMCID: PMC8151812 DOI: 10.3390/pathogens10050582] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/26/2021] [Accepted: 05/05/2021] [Indexed: 02/08/2023]
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2), has been recently considered a systemic disorder leading to the procoagulant state. Preliminary studies have shown that SARS-CoV-2 can infect endothelial cells, and extensive evidence of inflammation and endothelial dysfunction has been found in advanced COVID-19. Endothelial cells play a critical role in many physiological processes, such as controlling blood fluidity, leukocyte activation, adhesion, platelet adhesion and aggregation, and transmigration. Therefore, it is reasonable to think that endothelial dysfunction leads to vascular dysfunction, immune thrombosis, and inflammation associated with COVID-19. This article summarizes the association of endothelial dysfunction and SARS-CoV-2 infection and its therapeutic strategies.
Collapse
Affiliation(s)
- Hai Deng
- Division of Trauma & Surgical Critical Care, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (H.D.); (D.C.); (L.-S.T.)
| | - Ting-Xuan Tang
- Class 1901, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China;
| | - Deng Chen
- Division of Trauma & Surgical Critical Care, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (H.D.); (D.C.); (L.-S.T.)
| | - Liang-Sheng Tang
- Division of Trauma & Surgical Critical Care, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (H.D.); (D.C.); (L.-S.T.)
| | - Xiang-Ping Yang
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
| | - Zhao-Hui Tang
- Division of Trauma & Surgical Critical Care, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (H.D.); (D.C.); (L.-S.T.)
- Correspondence:
| |
Collapse
|
47
|
Marchese P, Lombardi M, Mantione ME, Baccellieri D, Ferrara D, Chiesa R, Alfieri O, Foglieni C. Confocal Blood Flow Videomicroscopy of Thrombus Formation over Human Arteries and Local Targeting of P2X7. Int J Mol Sci 2021; 22:ijms22084066. [PMID: 33920051 PMCID: PMC8071050 DOI: 10.3390/ijms22084066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 12/16/2022] Open
Abstract
Atherothrombosis exposes vascular components to blood. Currently, new antithrombotic therapies are emerging. Herein we investigated thrombogenesis of human arteries with/without atherosclerosis, and the interaction of coagulation and vascular components, we and explored the anti-thrombogenic efficacy of blockade of the P2X purinoceptor 7 (P2X7). A confocal blood flow videomicroscopy system was performed on cryosections of internal mammary artery (IMA) or carotid plaque (CPL) determining/localizing platelets and fibrin. Blood from healthy donors elicited thrombi over arterial layers. Confocal microscopy associated thrombus with tissue presence of collagen type I, laminin, fibrin(ogen) and tissue factor (TF). The addition of antibodies blocking TF (aTF) or factor XI (aFXI) to blood significantly reduced fibrin deposition, variable platelet aggregation and aTF + aFXI almost abolished thrombus formation, showing synergy between coagulation pathways. A scarce effect of aTF over sub-endothelial regions, more abundant in tissue TF and bundles of laminin and collagen type I than deep intima, may suggest tissue thrombogenicity as molecular structure-related. Consistently with TF-related vascular function and expression of P2X7, the sections from CPL but not IMA tissue cultures pre-treated with the P2X7 antagonist A740003 demonstrated poor thrombogenesis in flow experiments. These data hint to local targeting studies on P2X7 modulation for atherothrombosis prevention/therapy.
Collapse
Affiliation(s)
- Patrizia Marchese
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA;
| | - Maria Lombardi
- Myocardial Diseases and Atherosclerosis Unit, Cardiovascular Research Center, San Raffaele Scientific Institute IRCCS, Via Olgettina, 58, 20132 Milano, Italy; (M.L.); (M.E.M.)
| | - Maria Elena Mantione
- Myocardial Diseases and Atherosclerosis Unit, Cardiovascular Research Center, San Raffaele Scientific Institute IRCCS, Via Olgettina, 58, 20132 Milano, Italy; (M.L.); (M.E.M.)
| | - Domenico Baccellieri
- Cardiothoracic and Vascular Department, San Raffaele Scientific Institute IRCCS, Via Olgettina, 58, 20132 Milano, Italy; (D.B.); (D.F.); (R.C.); (O.A.)
| | - David Ferrara
- Cardiothoracic and Vascular Department, San Raffaele Scientific Institute IRCCS, Via Olgettina, 58, 20132 Milano, Italy; (D.B.); (D.F.); (R.C.); (O.A.)
| | - Roberto Chiesa
- Cardiothoracic and Vascular Department, San Raffaele Scientific Institute IRCCS, Via Olgettina, 58, 20132 Milano, Italy; (D.B.); (D.F.); (R.C.); (O.A.)
| | - Ottavio Alfieri
- Cardiothoracic and Vascular Department, San Raffaele Scientific Institute IRCCS, Via Olgettina, 58, 20132 Milano, Italy; (D.B.); (D.F.); (R.C.); (O.A.)
| | - Chiara Foglieni
- Myocardial Diseases and Atherosclerosis Unit, Cardiovascular Research Center, San Raffaele Scientific Institute IRCCS, Via Olgettina, 58, 20132 Milano, Italy; (M.L.); (M.E.M.)
- Correspondence: ; Tel.: +39-02-26434570
| |
Collapse
|
48
|
Enoxaparin prevents CXCL16/ADAM10-mediated cisplatin renal toxicity: Role of the coagulation system and the transcriptional factor NF-κB. Life Sci 2021; 270:119120. [PMID: 33545204 DOI: 10.1016/j.lfs.2021.119120] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/04/2021] [Accepted: 01/12/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND AIMS C-X-C ligand 16 (CXCL16) is an exceptional chemokine that is expressed as transmembrane and soluble forms. Our aim is to shed lights on the role of CXCL16/ADAM10 (a disintegrin and metalloproteinase) in cisplatin (CP)-induced renal toxicity as well as possible protective effect of enoxaparin. MAIN METHODS Male albino mice were injected with CP (30 mg/kg, i.p.) in the presence or absence of enoxaparin (ENOX) (5 mg/kg, i.p.). Renal toxicity markers, serum level of cystatin-c, complete blood count (CBC), prothrombin time (Pt) and tissue expression of CXCL16, ADAM10, cluster of differentiation 3 (CD3), fibrinogen, tissue factor (TF), nuclear factor-κB (NF-κB) and tumour necrosis factor α (TNF-α) were measured. Besides, serum CXCL16 and histopathology were also analyzed. KEY FINDINGS CP increased renal toxicity markers, renal expression of CXCL16/ADAM10, fibrinogen, TF and CD3 tissue expression in a time-dependent manner, and elevated serum cystatin-c, CXCL16 and tissue TNF-α, NF-κB. Alternatively, ENOX restored the deteriorated parameters and reduced tissue level of NF-κB. SIGNIFICANCE This report, for the first time, showed that soluble CXCL16 resulting from ADAM10 cleavage may recruit T-cells to the renal glomeruli and tubules in CP toxicity. Furthermore, TF and fibrin, have similar expression and location pattern like CXCL16 and ADAM10 suggesting their possible interrelation. ENOX successfully restored the deteriorated parameters suggesting it may be an effective nephroprotective adjuvant therapy.
Collapse
|
49
|
Kelham M, Choudry FA, Hamshere S, Beirne AM, Rathod KS, Baumbach A, Ahluwalia A, Mathur A, Jones DA. Therapeutic Implications of COVID-19 for the Interventional Cardiologist. J Cardiovasc Pharmacol Ther 2020; 26:203-216. [PMID: 33331160 DOI: 10.1177/1074248420982736] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Although COVID-19 is viewed primarily as a respiratory disease, cardiovascular risk factors and disease are prevalent among infected patients and are associated with worse outcomes. In addition, among multiple extra-pulmonary manifestations, there has been an increasing recognition of specific cardiovascular complications of COVID-19. Despite this, in the initial stages of the pandemic there was evidence of a reduction in patients presenting to acute cardiovascular services. In this masterclass review, with the aid of 2 exemplar cases, we will focus on the important therapeutic implications of COVID-19 for interventional cardiologists. We summarize the existing evidence base regarding the varied cardiovascular presentations seen in COVID-19 positive patients and the prognostic importance and potential mechanisms of acute myocardial injury in this setting. Importantly, through the use of a systematic review of the literature, we focus our discussion on the observed higher rates of coronary thrombus burden in patients with COVID-19 and acute coronary syndromes.
Collapse
Affiliation(s)
- Matthew Kelham
- Department of Cardiology, 560754Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom
| | - Fizzah A Choudry
- Department of Cardiology, 560754Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom.,Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, 4617Queen Mary University of London, London, United Kingdom
| | - Stephen Hamshere
- Department of Cardiology, 560754Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom
| | - Anne-Marie Beirne
- Department of Cardiology, 560754Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom.,Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, 4617Queen Mary University of London, London, United Kingdom
| | - Krishnaraj S Rathod
- Department of Cardiology, 560754Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom.,Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, 4617Queen Mary University of London, London, United Kingdom
| | - Andreas Baumbach
- Department of Cardiology, 560754Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom.,Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, 4617Queen Mary University of London, London, United Kingdom
| | - Amrita Ahluwalia
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, 4617Queen Mary University of London, London, United Kingdom
| | - Anthony Mathur
- Department of Cardiology, 560754Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom.,Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, 4617Queen Mary University of London, London, United Kingdom
| | - Daniel A Jones
- Department of Cardiology, 560754Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom.,Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, 4617Queen Mary University of London, London, United Kingdom
| |
Collapse
|
50
|
Wang Y, Shang W, Zhong H, Luo T, Niu M, Xu K, Tian J. Tumor Vessel Targeted Self-Assemble Nanoparticles for Amplification and Prediction of the Embolization Effect in Hepatocellular Carcinoma. ACS NANO 2020; 14:14907-14918. [PMID: 33111520 DOI: 10.1021/acsnano.0c04341] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Vessel embolization is recommended as the first line treatment for unresectable hepatocellular carcinoma (HCC). However, owing to the imprecise vessel embolization and heterogeneous response performance among patients, its survival benefits are often compromised. Herein, we reported an innovative strategy to extensively embolize the tumor by triggering the coagulation cascade, and predict the embolization effect with vessel density assessment. We synthesized manganese dioxide (MnO2)/verteporfin (BPD) nanocomposites, in which BPD bound to the tumor vessel endothelial cells (TVECs) and MnO2 nanosheets served as the carrier. MnO2 was reduced to Mn2+ ions and self-assembled with BPD to produce nanoBPD, resulting in enhanced TVECs apoptosis and coagulation cascade compared to that with free BPD. Furthermore, multimodal imaging was used to visualize tumor vessel density, which can be used as a predictor to identify the patients who would benefit from embolization. Our findings describe a promising strategy for both tumor eradication and effect prediction to improve survival benefits in unresectable HCC patients.
Collapse
Affiliation(s)
- Yaqin Wang
- Department of Interventional Radiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110000, China
| | - Wenting Shang
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Hongshan Zhong
- Department of Interventional Radiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110000, China
| | - Ting Luo
- Department of Interventional Radiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110000, China
| | - Meng Niu
- Department of Interventional Radiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110000, China
| | - Ke Xu
- Department of Interventional Radiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110000, China
| | - Jie Tian
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine, Beihang University, Beijing, 100191, China
| |
Collapse
|