1
|
Kostin S, Giannakopoulos T, Richter M, Krizanic F, Sasko B, Ritter O, Pagonas N. Coronary microthrombi in the failing human heart: the role of von Willebrand factor and PECAM-1. Mol Cell Biochem 2024; 479:3437-3446. [PMID: 38381272 PMCID: PMC11511743 DOI: 10.1007/s11010-024-04942-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/13/2024] [Indexed: 02/22/2024]
Abstract
The recognition of microthrombi in the heart microcirculation has recently emerged from studies in COVID-19 decedents. The present study investigated the ultrastructure of coronary microthrombi in heart failure (HF) due to cardiomyopathies that are unrelated to COVID-19 infection. In addition, we have investigated the role of von Willebrand factor (VWF) and PECAM-1 in microthrombus formation. We used electron microscopy to investigate the occurrence of microthrombi in patients with HF due to dilated (DCM, n = 7), inflammatory (MYO, n = 6) and ischemic (ICM, n = 7) cardiomyopathy and 4 control patients. VWF and PECAM-1 was studied by quantitative immunohistochemistry and Western blot. In comparison to control, the number of microthrombi was increased 7-9 times in HF. This was associated with a 3.5-fold increase in the number of Weibel-Palade bodies (WPb) in DCM and MYO compared to control. A fivefold increase in WPb in ICM was significantly different from control, DCM and MYO. In Western blot, VWF was increased twofold in DCM and MYO, and more than threefold in ICM. The difference between ICM and DCM and MYO was statistically significant. These results were confirmed by quantitative immunohistochemistry. Compared to control, PECAM-1 was by approximatively threefold increased in all groups of patients. This is the first study to demonstrate the occurrence of microthrombi in the failing human heart. The occurrence of microthrombi is associated with increased expression of VWF and the number of WPb, being more pronounced in ICM. These changes are likely not compensated by increases in PECAM-1 expression.
Collapse
Affiliation(s)
- Sawa Kostin
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany.
| | - Theodoros Giannakopoulos
- Department of Internal Medicine and Cardiology, Brandenburg Medical School Theodor Fontane, University Clinic Neuruppin-Brandenburg, Neuruppin, Germany
| | - Manfred Richter
- Department of Cardiac Surgery, Kerckhoff-Clinic, Bad Nauheim, Germany
| | - Florian Krizanic
- Department of Internal Medicine and Cardiology, Brandenburg Medical School Theodor Fontane, University Clinic Neuruppin-Brandenburg, Neuruppin, Germany
| | - Benjamin Sasko
- Medical Department II, Marien Hospital Herne, Ruhr-University of Bochum, Herne, Germany
| | - Oliver Ritter
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
- Department of Cardiology, University Hospital Brandenburg, Brandenburg an der Havel, Germany
| | - Nikolaos Pagonas
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Neuruppin, Germany
- Department of Internal Medicine and Cardiology, Brandenburg Medical School Theodor Fontane, University Clinic Neuruppin-Brandenburg, Neuruppin, Germany
| |
Collapse
|
2
|
Rehman M, Agarwal V, Chaudhary R, Kaushik AS, Srivastava S, Srivastava S, Kumar A, Singh S, Mishra V. Pharmacological inhibition of histone deacetylase alleviates chronic unpredictable stress induced atherosclerosis and endothelial dysfunction via upregulation of BDNF. Biochem Biophys Res Commun 2024; 735:150485. [PMID: 39098273 DOI: 10.1016/j.bbrc.2024.150485] [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: 06/02/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 08/06/2024]
Abstract
Long-term stress is a significant risk factor for cardiovascular diseases, including atherosclerosis and endothelial dysfunction. Moreover, prolonged stress has shown to negatively regulate central BDNF expression. The role of central BDNF in CNS disorders is well studied until recently the peripheral BDNF was also found to be involved in endothelial function regulation and atherosclerosis. The peripheral BDNF and its role in chronic stress-induced atherosclerosis and endothelial dysfunction remain unclear. Therefore, we aimed to elucidate the role of BDNF and its modulation by the HDAC inhibitor valproic acid (VA) in chronic unpredictable stress (CUS)-induced atherosclerosis and endothelial dysfunction. We demonstrated that a 10-week CUS mouse model substantially decreases central and peripheral BDNF expression, resulting in enhanced serum lipid indices, plaque deposition, fibrosis, and CD68 expression in thoracic aortas. Further, parameters associated with endothelial dysfunction such as increased levels of endothelin-1 (ET-1), adhesion molecules like VCAM-1, M1 macrophage markers, and decreased M2 macrophage markers, eNOS expression, and nitrite levels in aortas, were also observed. VA (50 mg/kg, 14 days, i. p.) was administered to mice following 8 weeks of CUS exposure until the end of the experimental procedure. VA significantly prevented the decrease in BDNF, eNOS and nitrite levels, reduced lesion formation and fibrosis in thoracic aortas and increased ET-1, and VCAM-1 followed by M2 polarization in VA-treated mice. The study highlights the potential of epigenetic modulation of BDNF as a therapeutic target, in stress-induced cardiovascular pathologies and suggests that VA could be a promising agent for mitigating CUS-induced endothelial dysfunction and atherosclerosis by BDNF modulation.
Collapse
Affiliation(s)
- Mujeeba Rehman
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh, 226025, India
| | - Vipul Agarwal
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh, 226025, India
| | - Rishabh Chaudhary
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh, 226025, India
| | - Arjun Singh Kaushik
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh, 226025, India
| | - Siddhi Srivastava
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh, 226025, India
| | - Sukriti Srivastava
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh, 226025, India
| | - Anand Kumar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh, 226025, India
| | - Sanjay Singh
- Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh, 226025, India.
| | - Vikas Mishra
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh, 226025, India.
| |
Collapse
|
3
|
Li RJ, Wen YX. A cross-sectional study of the association between blood cadmium and mortality among adults with myocardial infarction. Medicine (Baltimore) 2024; 103:e39705. [PMID: 39312320 PMCID: PMC11419487 DOI: 10.1097/md.0000000000039705] [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: 02/05/2024] [Accepted: 08/23/2024] [Indexed: 09/25/2024] Open
Abstract
Cadmium (Cd) plays a key role in the occurrence of myocardial infarction (MI). We aimed to explore the association between blood Cd levels and all-cause mortality of MI on the basis of the National Health and Nutrition Examination Survey databases. This study included 800 adults with MI to obtain blood Cd concentrations and their follow-up information. The association between Cd concentrations and mortality was analyzed using Cox regression, restricted cubic spline (RCS) models, mediation analysis, receiver operating characteristic curve, and Kaplan-Meier curves. All the patients were divided into 4 groups according to the quartiles of blood Cd levels (Q1, Q2, Q3, and Q4). Cox regression analysis with adjustment for covariates indicated that Cd was the promoting factor of mortality, and patients with higher Cd had a higher death risk. The RCS model indicated an "inverted checkmark" shaped correlation between Cd levels and mortality, and a turning point of 1.06 μg/L was found. A significant positive correlation was observed on the left of the turning point. Grouped patients by turning point into 2 groups, Kaplan-Meier analysis showed that the low-concentration group had a lower death risk than the high-concentration group. Subgroup analysis revealed that the prognostic effect of Cd was more pronounced in patients with former smoking history, and receiver operating characteristic curve showed that blood Cd had a better-predicting function in patients with MI. Blood Cd levels were significantly related to all-cause mortality in patients with MI, especially in patients with Cd < 1.06 μg/L.
Collapse
Affiliation(s)
- Ren-jie Li
- Emergency Department, Affiliated Hospital of Putian University, Putian, Fujian, China
| | - Ying-xu Wen
- Emergency Department, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| |
Collapse
|
4
|
Yang M, Boye-Doe A, Abosabie SAS, Barr AM, Mendez LM, Sharda AV. RalB uncoupled exocyst mediates endothelial Weibel-Palade body exocytosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.16.613344. [PMID: 39345530 PMCID: PMC11429928 DOI: 10.1101/2024.09.16.613344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Ras-like (Ral) GTPases play essential regulatory roles in many cellular processes, including exocytosis. Cycling between GDP- and GTP-bound states, Ral GTPases function as molecular switches and regulate effectors, specifically the multi-subunit tethering complex exocyst. Here, we show that Ral isoform RalB controls regulated exocytosis of Weibel-Palade bodies (WPBs), the specialized endothelial secretory granules that store hemostatic protein von Willebrand factor. Remarkably, unlike typical small GTPase-effector interactions, RalB binds exocyst in its GDP-bound state in resting endothelium. Upon endothelial cell stimulation, exocyst is uncoupled from RalB-GTP resulting in WPB tethering and exocytosis. Furthermore, we report that PKC-dependent phosphorylation of the C-terminal hypervariable region (HVR) of RalB modulates its dynamic interaction with exocyst in endothelium. Exocyst preferentially interacts with phosphorylated RalB in resting endothelium. Dephosphorylation of RalB either by endothelial cell stimulation, or PKC inhibition, or expression of nonphosphorylatable mutant at a specific serine residue of RalB HVR, disengages exocyst and augments WPB exocytosis, resembling RalB exocyst-binding site mutant. In summary, it is the uncoupling of exocyst from RalB that mediates endothelial Weibel-Palade body exocytosis. Our data shows that Ral function may be more dynamically regulated by phosphorylation and may confer distinct functionality given high degree of homology and the shared set of effector protein between the two Ral isoforms.
Collapse
Affiliation(s)
- Moua Yang
- Bloodworks Northwest Research Institute, Seattle, WA 98102, USA
- Division of Hematology and Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Alexandra Boye-Doe
- Section of Hematology, Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Salma A S Abosabie
- Section of Hematology, Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Alexandra M Barr
- Section of Hematology, Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Lourdes M Mendez
- Section of Hematology, Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Anish V Sharda
- Section of Hematology, Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
5
|
El-Mansi S, Mitchell TP, Mobayen G, McKinnon TAJ, Miklavc P, Frick M, Nightingale TD. Myosin-1C augments endothelial secretion of von Willebrand factor by linking contractile actomyosin machinery to the plasma membrane. Blood Adv 2024; 8:4714-4726. [PMID: 38669344 PMCID: PMC11413703 DOI: 10.1182/bloodadvances.2024012590] [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: 01/08/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
ABSTRACT Blood endothelial cells control the hemostatic and inflammatory response by secreting von Willebrand factor (VWF) and P-selectin from storage organelles called Weibel-Palade bodies (WPBs). Actin-associated motor proteins regulate this secretory pathway at multiple points. Before fusion, myosin Va forms a complex that anchors WPBs to peripheral actin structures, allowing for the maturation of content. After fusion, an actomyosin ring/coat is recruited and compresses the WPB to forcibly expel the largest VWF multimers. Here, we provide, to our knowledge, the first evidence for the involvement of class I myosins during regulated VWF secretion. We show that the unconventional myosin-1C (Myo1c) is recruited after fusion via its pleckstrin homology domain in an actin-independent process. This provides a link between the actin ring and phosphatidylinositol 4,5-bisphosphate (PIP2) at the membrane of the fused organelle and is necessary to ensure maximal VWF secretion. This is an active process requiring Myo1c ATPase activity because inhibition of class I myosins using the inhibitor pentachloropseudilin or expression of an ATPase-deficient Myo1c rigor mutant perturbs the expulsion of VWF and alters the kinetics of the exocytic actin ring. These data offer a novel insight into the control of an essential physiological process and provide a new way in which it can be regulated.
Collapse
Affiliation(s)
- Sammy El-Mansi
- Centre for Microvascular Research, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Tom P. Mitchell
- Centre for Microvascular Research, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Golzar Mobayen
- Department of Immunology and Inflammation, Centre for Haematology, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Thomas A. J. McKinnon
- Department of Immunology and Inflammation, Centre for Haematology, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Pika Miklavc
- School of Science, Engineering & Environment, University of Salford, Manchester, United Kingdom
| | - Manfred Frick
- Institute of General Physiology, Ulm University, Ulm, Germany
| | - Thomas D. Nightingale
- Centre for Microvascular Research, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
6
|
Hörber S, Prystupa K, Jacoby J, Fritsche A, Kleber ME, Moissl AP, Hellstern P, Peter A, März W, Wagner R, Heni M. Blood coagulation in Prediabetes clusters-impact on all-cause mortality in individuals undergoing coronary angiography. Cardiovasc Diabetol 2024; 23:306. [PMID: 39175055 PMCID: PMC11342575 DOI: 10.1186/s12933-024-02402-z] [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] [Received: 06/18/2024] [Accepted: 08/10/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND Metabolic clusters can stratify subgroups of individuals at risk for type 2 diabetes mellitus and related complications. Since obesity and insulin resistance are closely linked to alterations in hemostasis, we investigated the association between plasmatic coagulation and metabolic clusters including the impact on survival. METHODS Utilizing data from the Ludwigshafen Risk and Cardiovascular Health (LURIC) study, we assigned 917 participants without diabetes to prediabetes clusters, using oGTT-derived glucose and insulin, high-density lipoprotein cholesterol, triglycerides, and anthropometric data. We performed a comprehensive analysis of plasmatic coagulation parameters and analyzed their associations with mortality using proportional hazards models. Mediation analysis was performed to assess the effect of coagulation factors on all-cause mortality in prediabetes clusters. RESULTS Prediabetes clusters were assigned using published tools, and grouped into low-risk (clusters 1,2,4; n = 643) and high-risk (clusters 3,5,6; n = 274) clusters. Individuals in the high-risk clusters had a significantly increased risk of death (HR = 1.30; CI: 1.01 to 1.67) and showed significantly elevated levels of procoagulant factors (fibrinogen, FVII/VIII/IX), D-dimers, von-Willebrand factor, and PAI-1, compared to individuals in the low-risk clusters. In proportional hazards models adjusted for relevant confounders, elevated levels of fibrinogen, D-dimers, FVIII, and vWF were found to be associated with an increased risk of death. Multiple mediation analysis indicated that vWF significantly mediates the cluster-specific risk of death. CONCLUSIONS High-risk prediabetes clusters are associated with prothrombotic changes in the coagulation system that likely contribute to the increased mortality in those individuals at cardiometabolic risk. The hypercoagulable state observed in the high-risk clusters indicates an increased risk for cardiovascular and thrombotic diseases that should be considered in future risk stratification and therapeutic strategies.
Collapse
Affiliation(s)
- Sebastian Hörber
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, Tübingen, Germany.
- Institute of Diabetes Research and Metabolic Diseases, Helmholtz Center Munich German Research Center for Environmental Health, Tübingen, Germany.
- German Center for Diabetes Research, Neuherberg, Germany.
| | - Katsiaryna Prystupa
- German Center for Diabetes Research, Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
| | - Johann Jacoby
- Institute for Clinical Epidemiology and Applied Biometry, University Hospital Tübingen, Tübingen, Germany
| | - Andreas Fritsche
- Institute of Diabetes Research and Metabolic Diseases, Helmholtz Center Munich German Research Center for Environmental Health, Tübingen, Germany
- German Center for Diabetes Research, Neuherberg, Germany
- Department for Diabetology, Endocrinology, and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - Marcus E Kleber
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- SYNLAB MVZ für Humangenetik Mannheim GmbH, Mannheim, Germany
| | - Angela P Moissl
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Peter Hellstern
- Center of Hemostasis and Thrombosis Zurich, Zurich, Switzerland
| | - Andreas Peter
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, Tübingen, Germany
- Institute of Diabetes Research and Metabolic Diseases, Helmholtz Center Munich German Research Center for Environmental Health, Tübingen, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Winfried März
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- SYNLAB Academy, SYNLAB Holding Deutschland GmbH, Augsburg and Mannheim, Munich, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Robert Wagner
- German Center for Diabetes Research, Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany
| | - Martin Heni
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, Tübingen, Germany
- Division of Endocrinology and Diabetology, Department of Internal Medicine 1, University Hospital Ulm, Ulm, Germany
| |
Collapse
|
7
|
Smith DR, Lim ST, Murphy SJX, Hickey FB, Offiah C, Murphy SM, Collins DR, Coughlan T, O'Neill D, Egan B, O'Donnell JS, O'Sullivan JM, McCabe DJH. von Willebrand factor antigen, von Willebrand factor propeptide and ADAMTS13 activity in TIA or ischaemic stroke patients changing antiplatelet therapy. J Neurol Sci 2024; 463:123118. [PMID: 39024743 DOI: 10.1016/j.jns.2024.123118] [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: 03/29/2024] [Revised: 06/10/2024] [Accepted: 06/28/2024] [Indexed: 07/20/2024]
Abstract
Data are limited on the impact of commencing antiplatelet therapy on von Willebrand Factor Antigen (VWF:Ag) or von Willebrand Factor propeptide (VWFpp) levels and ADAMTS13 activity, and their relationship with platelet reactivity following TIA/ischaemic stroke. In this pilot, observational study, VWF:Ag and VWFpp levels and ADAMTS13 activity were quantified in 48 patients ≤4 weeks of TIA/ischaemic stroke (baseline), and 14 days (14d) and 90 days (90d) after commencing aspirin, clopidogrel or aspirin+dipyridamole. Platelet reactivity was assessed at moderately-high shear stress (PFA-100® Collagen-Epinephrine / Collagen-ADP / INNOVANCE PFA P2Y assays), and low shear stress (VerifyNow® Aspirin / P2Y12, and Multiplate® Aspirin / ADP assays). VWF:Ag levels decreased and VWFpp/VWF:Ag ratio increased between baseline and 14d and 90d in the overall population (P ≤ 0.03). In the clopidogrel subgroup, VWF:Ag levels decreased and VWFpp/VWF:Ag ratio increased between baseline and 14d and 90d (P ≤ 0.01), with an increase in ADAMTS13 activity between baseline vs. 90d (P ≤ 0.03). In the aspirin+dipyridamole subgroup, there was an inverse relationship between VWF:Ag and VWFpp levels with both PFA-100 C-ADP and INNOVANCE PFA P2Y closure times (CTs) at baseline (P ≤ 0.02), with PFA-100 C-ADP, INNOVANCE PFA P2Y and C-EPI CTs at 14d (P ≤ 0.05), and between VWF:Ag levels and PFA-100 INNOVANCE PFA P2Y CTs at 90d (P = 0.03). There was a positive relationship between ADAMTS13 activity and PFA-100 C-ADP CTs at baseline (R2 = 0.254; P = 0.04). Commencing/altering antiplatelet therapy, mainly attributed to commencing clopidogrel in this study, was associated with decreasing endothelial activation following TIA/ischaemic stroke. These data enhance our understanding of the impact of VWF:Ag and VWFpp especially on ex-vivo platelet reactivity status at high shear stress after TIA/ischaemic stroke.
Collapse
Affiliation(s)
- D R Smith
- Vascular Neurology Research Foundation, c/o Department of Neurology, Tallaght University Hospital (TUH) / The Adelaide and Meath Hospital, Dublin, incorporating the National Children's Hospital (AMNCH), Dublin, Ireland; Department of Neurology, TUH / AMNCH, Dublin, Ireland; Academic Unit of Neurology, School of Medicine, Trinity College Dublin, Ireland
| | - S T Lim
- Department of Neurology, TUH / AMNCH, Dublin, Ireland; Stroke Service, TUH / AMNCH, Dublin, Ireland; Department of Clinical and Movement Neurosciences, Royal Free Campus, UCL Queen Square Institute of Neurology, London, UK; Academic Unit of Neurology, School of Medicine, Trinity College Dublin, Ireland
| | - S J X Murphy
- Department of Neurology, TUH / AMNCH, Dublin, Ireland; Stroke Service, TUH / AMNCH, Dublin, Ireland; Academic Unit of Neurology, School of Medicine, Trinity College Dublin, Ireland
| | - F B Hickey
- Trinity Centre for Health Sciences, Dept. of Clinical Medicine, School of Medicine, Trinity College Dublin, Ireland
| | - C Offiah
- Department of Neurology, TUH / AMNCH, Dublin, Ireland; Stroke Service, TUH / AMNCH, Dublin, Ireland; Academic Unit of Neurology, School of Medicine, Trinity College Dublin, Ireland
| | - S M Murphy
- Department of Neurology, TUH / AMNCH, Dublin, Ireland; Stroke Service, TUH / AMNCH, Dublin, Ireland; Academic Unit of Neurology, School of Medicine, Trinity College Dublin, Ireland
| | - D R Collins
- Department of Age-Related Health Care, TUH / AMNCH, Dublin, Ireland; Stroke Service, TUH / AMNCH, Dublin, Ireland
| | - T Coughlan
- Department of Age-Related Health Care, TUH / AMNCH, Dublin, Ireland; Stroke Service, TUH / AMNCH, Dublin, Ireland
| | - D O'Neill
- Department of Age-Related Health Care, TUH / AMNCH, Dublin, Ireland; Stroke Service, TUH / AMNCH, Dublin, Ireland
| | - B Egan
- Department of Vascular Surgery, TUH / AMNCH, Dublin, Ireland
| | - J S O'Donnell
- National Coagulation Centre, St James's Hospital, Dublin, Ireland; Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - J M O'Sullivan
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - D J H McCabe
- Vascular Neurology Research Foundation, c/o Department of Neurology, Tallaght University Hospital (TUH) / The Adelaide and Meath Hospital, Dublin, incorporating the National Children's Hospital (AMNCH), Dublin, Ireland; Department of Neurology, TUH / AMNCH, Dublin, Ireland; Stroke Service, TUH / AMNCH, Dublin, Ireland; Department of Clinical and Movement Neurosciences, Royal Free Campus, UCL Queen Square Institute of Neurology, London, UK; Academic Unit of Neurology, School of Medicine, Trinity College Dublin, Ireland.
| |
Collapse
|
8
|
Sarkar A, Fanous KI, Marei I, Ding H, Ladjimi M, MacDonald R, Hollenberg MD, Anderson TJ, Hill MA, Triggle CR. Repurposing Metformin for the Treatment of Atrial Fibrillation: Current Insights. Vasc Health Risk Manag 2024; 20:255-288. [PMID: 38919471 PMCID: PMC11198029 DOI: 10.2147/vhrm.s391808] [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] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 06/05/2024] [Indexed: 06/27/2024] Open
Abstract
Metformin is an orally effective anti-hyperglycemic drug that despite being introduced over 60 years ago is still utilized by an estimated 120 to 150 million people worldwide for the treatment of type 2 diabetes (T2D). Metformin is used off-label for the treatment of polycystic ovary syndrome (PCOS) and for pre-diabetes and weight loss. Metformin is a safe, inexpensive drug with side effects mostly limited to gastrointestinal issues. Prospective clinical data from the United Kingdom Prospective Diabetes Study (UKPDS), completed in 1998, demonstrated that metformin not only has excellent therapeutic efficacy as an anti-diabetes drug but also that good glycemic control reduced the risk of micro- and macro-vascular complications, especially in obese patients and thereby reduced the risk of diabetes-associated cardiovascular disease (CVD). Based on a long history of clinical use and an excellent safety record metformin has been investigated to be repurposed for numerous other diseases including as an anti-aging agent, Alzheimer's disease and other dementias, cancer, COVID-19 and also atrial fibrillation (AF). AF is the most frequently diagnosed cardiac arrythmia and its prevalence is increasing globally as the population ages. The argument for repurposing metformin for AF is based on a combination of retrospective clinical data and in vivo and in vitro pre-clinical laboratory studies. In this review, we critically evaluate the evidence that metformin has cardioprotective actions and assess whether the clinical and pre-clinical evidence support the use of metformin to reduce the risk and treat AF.
Collapse
Affiliation(s)
- Aparajita Sarkar
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Kareem Imad Fanous
- Department of Medical Education, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Isra Marei
- Department of Pharmacology & Medical Education, Weill Cornell Medicine- Qatar, Doha, Qatar
| | - Hong Ding
- Department of Pharmacology & Medical Education, Weill Cornell Medicine- Qatar, Doha, Qatar
| | - Moncef Ladjimi
- Department of Biochemistry & Medical Education, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Ross MacDonald
- Health Sciences Library, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Morley D Hollenberg
- Department of Physiology & Pharmacology, and Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Todd J Anderson
- Department of Cardiac Sciences and Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Michael A Hill
- Dalton Cardiovascular Research Center & Department of Medical Pharmacology & Physiology, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Chris R Triggle
- Department of Pharmacology & Medical Education, Weill Cornell Medicine- Qatar, Doha, Qatar
| |
Collapse
|
9
|
Gakuba C, Dumitrascu AD, Marsan PE, Legallois D, Hanouz JL, Vivien D, Martinez de Lizarrondo S, Gauberti M, Cerasuolo D. N-Acetylcysteine to Reduce Mortality for Patients Requiring Cardiac Catheterization or Cardiac Surgery: A Systematic Review and Meta-analysis. J Cardiovasc Pharmacol 2024; 83:580-587. [PMID: 38467037 DOI: 10.1097/fjc.0000000000001551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 01/31/2024] [Indexed: 03/13/2024]
Abstract
ABSTRACT Multimers of von Willebrand factor play a critical role in various processes inducing morbidity and mortality in cardiovascular-risk patients. With the ability to reduce von Willebrand factor multimers, N-acetylcysteine (NAC) could reduce mortality in patients undergoing coronary catheterization or cardiac surgery. However, its impact in perioperative period has never been studied so far in regard of its potential cardiovascular benefits. Then, 4 databases were searched for randomized controlled trials that compared in-hospital mortality between an experimental group, with NAC, and a control group without NAC, in patients undergoing coronary catheterization or cardiac surgery. The primary efficacy outcome was in-hospital mortality. Secondary outcomes were the occurrence of thrombotic events, major cardiovascular events, myocardial infarction, and contrast-induced nephropathy. The safety outcome was occurrence of hemorrhagic events. Nineteen studies totaling 3718 patients were included. Pooled analysis demonstrated a reduction of in-hospital mortality associated with NAC: odds ratio, 0.60; 95% confidence interval, 0.39-0.92; P = 0.02. The occurrence of secondary outcomes was not significantly reduced with NAC except for contrast-induced nephropathy. No difference was reported for hemorrhagic events. Subgroup analyses revealed a life-saving effect of NAC in a dose-dependent manner with reduction of in-hospital mortality for the NAC high-dose group, but not for the NAC standard-dose (<3500-mg) group. In conclusion, without being able to conclude on the nature of the mechanism involved, our review suggests a benefit of NAC in cardiovascular-risk patients in perioperative period in terms of mortality and supports prospective confirmatory studies.
Collapse
Affiliation(s)
- Clement Gakuba
- département d'Anesthésie Réanimation, CHU de Caen Normandie, Caen, France
- département PhIND « Physiopathology and imaging of Neurological Disorders », Institut Blood and Brain @ Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen, France
| | | | | | - Damien Legallois
- département de Cardiologie, CHU de Caen Normandie, EA4650 (SEILIRM), FHU REMOD-VHF,Caen, France
| | - Jean-Luc Hanouz
- département d'Anesthésie Réanimation, CHU de Caen Normandie, Caen, France
- département PhIND « Physiopathology and imaging of Neurological Disorders », Institut Blood and Brain @ Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen, France
| | - Denis Vivien
- département PhIND « Physiopathology and imaging of Neurological Disorders », Institut Blood and Brain @ Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen, France
- département de Recherche Clinique, CHU de Caen Normandie, Caen, France
| | - Sara Martinez de Lizarrondo
- département PhIND « Physiopathology and imaging of Neurological Disorders », Institut Blood and Brain @ Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen, France
| | - Maxime Gauberti
- département PhIND « Physiopathology and imaging of Neurological Disorders », Institut Blood and Brain @ Caen-Normandie, Normandie Univ, UNICAEN, INSERM, U1237, Cyceron, Caen, France
- département de Radiologie diagnostique et interventionnelle, CHU de Caen Normandie, Caen, France
| | - Damiano Cerasuolo
- département de santé publique, Centre hospitalier universitaire de Caen, Caen, France; and
- Normandie Univ, UNICAEN, INSERM U1086, ANTICIPE, Caen, France
| |
Collapse
|
10
|
Pérez-Jurado LA, Cáceres A, Balagué-Dobón L, Esko T, López de Heredia M, Quintela I, Cruz R, Lapunzina P, Carracedo Á, González JR. Clonal chromosomal mosaicism and loss of chromosome Y in elderly men increase vulnerability for SARS-CoV-2. Commun Biol 2024; 7:202. [PMID: 38374351 PMCID: PMC10876565 DOI: 10.1038/s42003-024-05805-6] [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: 02/18/2022] [Accepted: 01/11/2024] [Indexed: 02/21/2024] Open
Abstract
The pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2, COVID-19) had an estimated overall case fatality ratio of 1.38% (pre-vaccination), being 53% higher in males and increasing exponentially with age. Among 9578 individuals diagnosed with COVID-19 in the SCOURGE study, we found 133 cases (1.42%) with detectable clonal mosaicism for chromosome alterations (mCA) and 226 males (5.08%) with acquired loss of chromosome Y (LOY). Individuals with clonal mosaic events (mCA and/or LOY) showed a 54% increase in the risk of COVID-19 lethality. LOY is associated with transcriptomic biomarkers of immune dysfunction, pro-coagulation activity and cardiovascular risk. Interferon-induced genes involved in the initial immune response to SARS-CoV-2 are also down-regulated in LOY. Thus, mCA and LOY underlie at least part of the sex-biased severity and mortality of COVID-19 in aging patients. Given its potential therapeutic and prognostic relevance, evaluation of clonal mosaicism should be implemented as biomarker of COVID-19 severity in elderly people.
Collapse
Affiliation(s)
- Luis A Pérez-Jurado
- Genetics Unit, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain.
- Genetics Service, Hospital del Mar & Hospital del Mar Research Institute (IMIM), Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Barcelona, Spain.
| | - Alejandro Cáceres
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain
- Centro de Investigación Biomédica en Red en Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Laura Balagué-Dobón
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain
- Centro de Investigación Biomédica en Red en Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Tonu Esko
- Estonian Genome Science Centre, University of Tartu, Tartu, Estonia
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Miguel López de Heredia
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Barcelona, Spain
| | - Inés Quintela
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Barcelona, Spain
- Centro Nacional de Genotipado (CEGEN), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Raquel Cruz
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Barcelona, Spain
- Centro Nacional de Genotipado (CEGEN), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
- Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Pablo Lapunzina
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Barcelona, Spain
- Instituto de Genética Médica y Molecular (INGEMM), Hospital Universitario La Paz-IDIPAZ, Madrid, Spain
- ERN-ITHACA-European Reference Network, Paris, France
| | - Ángel Carracedo
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Barcelona, Spain
- Centro Nacional de Genotipado (CEGEN), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
- Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Fundación Pública Galega de Medicina Xenómica, Sistema Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Juan R González
- Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain.
- Centro de Investigación Biomédica en Red en Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain.
- Department of Mathematics, Universitat Autònoma de Barcelona, Bellaterra, Spain.
| |
Collapse
|
11
|
Cox AA, Liu A, Ng CJ. Clusterin knockdown has effects on intracellular and secreted von Willebrand factor in human umbilical vein endothelial cells. PLoS One 2024; 19:e0298133. [PMID: 38363768 PMCID: PMC10871512 DOI: 10.1371/journal.pone.0298133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 01/17/2024] [Indexed: 02/18/2024] Open
Abstract
Alterations in von Willebrand factor (VWF) have an important role in human health and disease. Deficiency of VWF is associated with symptoms of bleeding and excesses of VWF are associated with thrombotic outcomes. Understanding the mechanisms that drive VWF regulation can lead to a better understanding of modulation of VWF levels in humans. We identified clusterin (CLU) as a potential candidate regulator of VWF based on a single cell RNA sequencing (scRNA-seq) analysis in control endothelial cells (ECs) and von Willebrand disease (VWD) endothelial colony-forming-cells (ECFCs). We found that patients with deficiencies of VWF (von Willebrand disease, VWD) had decreased CLU expression and ECs with low VWF expression also had low CLU expression. Based on these findings, we sought to evaluate the role of CLU in the regulation of VWF, specifically as it relates to VWD. As CLU is primarily thought to be a golgi protein involved in protein chaperoning, we hypothesized that knockdown of CLU would lead to decreases in VWF and alterations in Weibel-Palade bodies (WPBs). We used both siRNA- and CRISPR-Cas9-based approaches to modulate CLU in human umbilical vein endothelial cells (HUVECs) and evaluated VWF protein levels, VWF mRNA copy number, and WPB quantity and size. We demonstrated that siRNA-based knockdown of CLU resulted in decreases in VWF content in cellular lysates and supernatants, but no significant change in WPB quantity or size. A CRISPR-Cas9-based knockdown of CLU demonstrated similar findings of decreases in intracellular VWF content but no significant change in WPB quantity or size. Our data suggests that CLU knockdown is associated with decreases in cellular VWF content but does not affect VWF mRNA levels or WPB quantity or size.
Collapse
Affiliation(s)
- Allaura A. Cox
- Department of Pediatrics, University of Colorado–Anschutz Medical Campus, Aurora, CO, United States of America
| | - Alice Liu
- Department of Bioengineering, Washington University, St. Louis, MO, United States of America
| | - Christopher J. Ng
- Department of Pediatrics, University of Colorado–Anschutz Medical Campus, Aurora, CO, United States of America
| |
Collapse
|
12
|
Keret S, Mazareeb J, Snir A, Shouval A, Awisat A, Kaly L, Rosner I, Rozenbaum M, Boulman N, Hardak E, Slobodin G, Rimar D. Von Willebrand factor: a possible biomarker for disease activity in vasculitis. Scand J Rheumatol 2024:1-9. [PMID: 38314784 DOI: 10.1080/03009742.2024.2302679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 01/04/2024] [Indexed: 02/07/2024]
Abstract
OBJECTIVE Inflammation markers, e.g. C- reactive protein (CRP) and sedimentation rate, can be normal despite active vasculitis. Von Willebrand factor (vWF) is secreted from endothelial cells in response to vascular damage. Some reports suggest increased vWF levels in vasculitis. This study aimed to evaluate vWF serum concentration in vasculitis patients as a possible biomarker of disease activity and to review the current literature. METHOD Adult patients with systemic vasculitis were prospectively enrolled. Disease activity was recorded using the Birmingham Vasculitis Activity Score (BVAS) version 3. Blood group-adjusted vWF antigen serum level was evaluated at diagnosis and, when available, after treatment. RESULTS Twenty-five vasculitis patients were compared to 15 healthy controls. The mean age of patients was 56 ± 17 years and 56% were women. Forty percent had anti-neutrophil cytoplasmic autoantibody-associated vasculitis, 20% giant cell arteritis, 16% polyarteritis nodosa, 8% Takayasu arteritis, and the rest had other vasculitides. The mean disease duration was 3.4 ± 4.8 years. Mean vWF was higher in patients with active vasculitis than in healthy controls (212 ± 81% vs 106 ± 26%, p < 0.001). vWF levels directly correlated with BVAS. In 13 patients with active vasculitis who reached remission or low disease activity after treatment, vWF level at follow-up decreased significantly. In three out of five patients who were treated with interleukin-6 inhibitors, vWF was elevated despite normal CRP levels, while vasculitis was clinically active. CONCLUSION vWF antigen serum level is increased in active vasculitis and could potentially serve as a biomarker for active disease.
Collapse
Affiliation(s)
- S Keret
- Rheumatology Unit, Bnai Zion Medical Center, Faculty of Medicine, Technion, Haifa, Israel
| | - J Mazareeb
- Rheumatology Unit, Bnai Zion Medical Center, Faculty of Medicine, Technion, Haifa, Israel
| | - A Snir
- Hematology Laboratory, Bnai Zion Medical Center, Haifa, Israel
| | - A Shouval
- Rheumatology Unit, Bnai Zion Medical Center, Faculty of Medicine, Technion, Haifa, Israel
| | - A Awisat
- Rheumatology Unit, Bnai Zion Medical Center, Faculty of Medicine, Technion, Haifa, Israel
| | - L Kaly
- Rheumatology Unit, Bnai Zion Medical Center, Faculty of Medicine, Technion, Haifa, Israel
| | - I Rosner
- Rheumatology Unit, Bnai Zion Medical Center, Faculty of Medicine, Technion, Haifa, Israel
| | - M Rozenbaum
- Rheumatology Unit, Bnai Zion Medical Center, Faculty of Medicine, Technion, Haifa, Israel
| | - N Boulman
- Rheumatology Unit, Bnai Zion Medical Center, Faculty of Medicine, Technion, Haifa, Israel
| | - E Hardak
- Pulmonary Unit, Bnai Zion Medical Center, Faculty of Medicine, Technion, Haifa, Israel
| | - G Slobodin
- Rheumatology Unit, Bnai Zion Medical Center, Faculty of Medicine, Technion, Haifa, Israel
| | - D Rimar
- Rheumatology Unit, Bnai Zion Medical Center, Faculty of Medicine, Technion, Haifa, Israel
| |
Collapse
|
13
|
Rafaqat S, Gluscevic S, Patoulias D, Sharif S, Klisic A. The Association between Coagulation and Atrial Fibrillation. Biomedicines 2024; 12:274. [PMID: 38397876 PMCID: PMC10887311 DOI: 10.3390/biomedicines12020274] [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: 12/15/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
The existing literature highlights the presence of numerous coagulation factors and markers. Elevated levels of coagulation factors are associated with both existing and newly diagnosed cases of atrial fibrillation (AF). However, this article summarizes the role of coagulation in the pathogenesis of AF, which includes fibrinogen and fibrin, prothrombin, thrombomodulin, soluble urokinase plasminogen activator receptor, von Willebrand factor, P-selectin, D-dimer, plasminogen activator inhibitor-1, and platelet activation. Coagulation irregularities play a significant role in the pathogenesis of AF.
Collapse
Affiliation(s)
- Saira Rafaqat
- Department of Zoology (Molecular Physiology), Lahore College for Women University, Lahore 54600, Punjab, Pakistan
| | - Sanja Gluscevic
- Department of Neurology, Clinical Center of Montenegro, 81000 Podgorica, Montenegro
| | - Dimitrios Patoulias
- Outpatient Department of Cardiometabolic Medicine, Second Department of Cardiology, Aristotle University of Thessaloniki, General Hospital “Hippokration”, 54642 Thessaloniki, Greece
| | - Saima Sharif
- Department of Zoology (Molecular Physiology), Lahore College for Women University, Lahore 54600, Punjab, Pakistan
| | - Aleksandra Klisic
- Faculty of Medicine, University of Montenegro, 81000 Podgorica, Montenegro
- Center for Laboratory Diagnostics, Primary Health Care Center, 81000 Podgorica, Montenegro
| |
Collapse
|
14
|
Košuta D, Novaković M, Božič Mijovski M, Jug B. Acute effects of high intensity interval training versus moderate intensity continuous training on haemostasis in patients with coronary artery disease. Sci Rep 2024; 14:1963. [PMID: 38263210 PMCID: PMC10806221 DOI: 10.1038/s41598-024-52521-6] [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: 10/08/2023] [Accepted: 01/19/2024] [Indexed: 01/25/2024] Open
Abstract
Exercise training is associated with an acute net increase in coagulation, which may increase the risk of atherothrombosis in coronary artery disease (CAD) patients. We sought to compare the acute haemostatic effects of a bout of moderate-intensity continuous (MICT) and high-intensity interval training (HIIT) in patients with CAD. Patients after a recent myocardial infarction were randomized into a HIIT or MICT session of exercise training on a stationary bike. Blood was sampled at baseline, after the exercise bout and after a one-hour resting period. We measured overall haemostatic potential (OHP), overall coagulation potential (OCP), fibrinogen, D-dimer and von Willebrand factor (vWF) and calculated overall fibrinolytic potential (OFP). Linear mixed models for repeated measures were constructed to assess the treatment effect. A total of 117 patients were included. OCP, OHP, fibrinogen, D-dimer and vWF significantly increased after exercise and returned to baseline after a one-hour rest, OFP decreased after exercise and returned to baseline levels after a one-hour rest. Linear mixed models showed a significant difference between HIIT and MICT in fibrinogen (p 0.043) and D-dimer (p 0.042). Our study has shown that an exercise bout is associated with a transient procoagulant state in patients with CAD, with similar exercise-induced haemostatic changes for HIIT and MICT.
Collapse
Affiliation(s)
- Daniel Košuta
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
- Department of Vascular Diseases, University Medical Centre Ljubljana, Zaloška Cesta 7, 1000, Ljubljana, Slovenia.
| | - Marko Novaković
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Vascular Diseases, University Medical Centre Ljubljana, Zaloška Cesta 7, 1000, Ljubljana, Slovenia
| | - Mojca Božič Mijovski
- Department of Vascular Diseases, University Medical Centre Ljubljana, Zaloška Cesta 7, 1000, Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Borut Jug
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Vascular Diseases, University Medical Centre Ljubljana, Zaloška Cesta 7, 1000, Ljubljana, Slovenia
| |
Collapse
|
15
|
Casella C, Kiles F, Urquhart C, Michaud DS, Kirwa K, Corlin L. Methylomic, Proteomic, and Metabolomic Correlates of Traffic-Related Air Pollution in the Context of Cardiorespiratory Health: A Systematic Review, Pathway Analysis, and Network Analysis. TOXICS 2023; 11:1014. [PMID: 38133415 PMCID: PMC10748071 DOI: 10.3390/toxics11121014] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/18/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023]
Abstract
A growing body of literature has attempted to characterize how traffic-related air pollution (TRAP) affects molecular and subclinical biological processes in ways that could lead to cardiorespiratory disease. To provide a streamlined synthesis of what is known about the multiple mechanisms through which TRAP could lead to cardiorespiratory pathology, we conducted a systematic review of the epidemiological literature relating TRAP exposure to methylomic, proteomic, and metabolomic biomarkers in adult populations. Using the 139 papers that met our inclusion criteria, we identified the omic biomarkers significantly associated with short- or long-term TRAP and used these biomarkers to conduct pathway and network analyses. We considered the evidence for TRAP-related associations with biological pathways involving lipid metabolism, cellular energy production, amino acid metabolism, inflammation and immunity, coagulation, endothelial function, and oxidative stress. Our analysis suggests that an integrated multi-omics approach may provide critical new insights into the ways TRAP could lead to adverse clinical outcomes. We advocate for efforts to build a more unified approach for characterizing the dynamic and complex biological processes linking TRAP exposure and subclinical and clinical disease and highlight contemporary challenges and opportunities associated with such efforts.
Collapse
Affiliation(s)
- Cameron Casella
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA; (C.C.); (F.K.); (C.U.); (D.S.M.); (K.K.)
| | - Frances Kiles
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA; (C.C.); (F.K.); (C.U.); (D.S.M.); (K.K.)
| | - Catherine Urquhart
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA; (C.C.); (F.K.); (C.U.); (D.S.M.); (K.K.)
| | - Dominique S. Michaud
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA; (C.C.); (F.K.); (C.U.); (D.S.M.); (K.K.)
| | - Kipruto Kirwa
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA; (C.C.); (F.K.); (C.U.); (D.S.M.); (K.K.)
- Department of Environmental Health, Boston University School of Public Health, Boston, MA 02118, USA
| | - Laura Corlin
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA; (C.C.); (F.K.); (C.U.); (D.S.M.); (K.K.)
- Department of Civil and Environmental Engineering, Tufts University School of Engineering, Medford, MA 02155, USA
| |
Collapse
|
16
|
Ryu JH, Bauer KA, Schulman S. Periprocedural management of type 2N von Willebrand disease with efanesoctocog alfa. J Thromb Haemost 2023; 21:3508-3510. [PMID: 37734716 DOI: 10.1016/j.jtha.2023.09.009] [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: 09/05/2023] [Accepted: 09/11/2023] [Indexed: 09/23/2023]
Abstract
Type 2 Normandy von Willebrand disease (type 2N VWD) is a rare qualitative defect in von Willebrand factor (VWF) that results in impaired factor VIII (FVIII) binding and consequently reduced FVIII levels. Current perioperative strategies require VWF concentrates to attain durable hemostatic FVIII levels. This case highlights the successful perioperative management of a 78-year-old female with type 2N VWD and coronary artery disease utilizing efanesoctocog alfa, a novel long-acting recombinant FVIII product approved for hemophilia A. By decoupling the FVIII-VWF interaction, efanesoctocog alfa achieves prolonged FVIII circulation independent of VWF. A single administration targeting 90% FVIII levels yielded sustained FVIII elevation without achieving supraphysiologic VWF levels, thus mitigating potential cardiovascular risks. This is the first report of efanesoctocog alfa use in type 2N VWD. Further clinical studies are necessary to corroborate its efficacy and safety for this indication.
Collapse
Affiliation(s)
- Justine H Ryu
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA; Division of Hematology and Hematologic Malignancies, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Kenneth A Bauer
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA; Division of Hematology and Hematologic Malignancies, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Sol Schulman
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA; Division of Hematology and Hematologic Malignancies, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
17
|
van den Boom BP, Stamouli M, Timon J, Bernal W, Blasi A, Adelmeijer J, Fernandez J, Lisman T, Patel VC. Von Willebrand factor is an independent predictor of short-term mortality in acutely ill patients with cirrhosis. Liver Int 2023; 43:2752-2761. [PMID: 37715606 DOI: 10.1111/liv.15728] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 07/25/2023] [Accepted: 08/30/2023] [Indexed: 09/17/2023]
Abstract
BACKGROUND AND AIMS Levels of von Willebrand factor (VWF) are elevated in patients with cirrhosis, and correlate well with disease severity. In patients with decompensated cirrhosis (DC), plasma VWF is associated with mortality. The value of VWF in predicting short-term mortality risk in patients with acute-on-chronic liver failure (ACLF) is, however, unclear. METHODS We included patients with DC (n = 111) and ACLF (n = 105). We measured VWF levels and correlated these with other laboratory parameters and prediction models for mortality. Also, we assessed the predictive value of VWF in the prediction of 90- and 30-day mortality in patients with DC and ACLF, respectively, and compared this to the predictive value of clinically used prediction models. Finally, we determined the optimal cut-off value for VWF in patients with ACLF. RESULTS Sixteen of 111 (14%) patients with DC and 35 of 105 (33%) with ACLF died within 90 and 30 days, respectively. VWF was associated with mortality and correlated closely with other prediction models. In patients with ACLF, VWF levels had a discrimination for 30-day mortality comparable with these models and accurately identified ACLF patients with high 30-day mortality risk. CONCLUSIONS Levels of VWF associate closely with risk of mortality in patients with DC and ACLF, and may have predictive utility as a laboratory marker of prognosis. Further research is warranted to assess the additional value of VWF in the prediction of mortality and associated complications in chronic liver failure syndromes.
Collapse
Affiliation(s)
- Bente P van den Boom
- Surgical Research Laboratory and Section of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Marilena Stamouli
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London, UK
| | - Jennifer Timon
- Institute of Liver Studies & Transplantation, King's College Hospital, NHS Foundation Trust, London, UK
| | - William Bernal
- Institute of Liver Studies & Transplantation, King's College Hospital, NHS Foundation Trust, London, UK
| | - Annabel Blasi
- Anesthesia Department, Hospital Clinic, IDIBAPS, Barcelona, Spain
| | - Jelle Adelmeijer
- Surgical Research Laboratory and Section of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Ton Lisman
- Surgical Research Laboratory and Section of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Vishal C Patel
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London, UK
- Institute of Liver Studies & Transplantation, King's College Hospital, NHS Foundation Trust, London, UK
- Liver Sciences, School of Immunology & Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| |
Collapse
|
18
|
Shaverdian M, Li RHL. Preventing Cardiogenic Thromboembolism in Cats: Literature Gaps, Rational Recommendations, and Future Therapies. Vet Clin North Am Small Anim Pract 2023; 53:1309-1323. [PMID: 37516545 DOI: 10.1016/j.cvsm.2023.06.002] [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] [Indexed: 07/31/2023]
Abstract
Cardiogenic arterial thromboembolism (CATE) is a devastating complication in cats with cardiomyopathies with significant morbidity and mortality. Despite recent advances in the diagnosis and treatment of CATE, its recurrence and mortality remain high. This highlights the urgent need for a greater understanding of CATE pathophysiology so that novel diagnostic tests and therapeutics can be developed. This comprehensive review aims to summarize existing literature on pathophysiology, clinical diagnosis, and current recommendations on the prevention and treatment of CATE. It also identifies and describes knowledge gaps and research priorities in the roles of immunothrombosis and procoagulant platelets in the pathogenesis of CATE.
Collapse
Affiliation(s)
- Meg Shaverdian
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, 2108 Tupper Hall, One Shields Avenue, Davis, CA 95616, USA
| | - Ronald H L Li
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA.
| |
Collapse
|
19
|
Alavi P, Yousef Abdualla R, Brown D, Mojiri A, Nagendran J, Lewis J, Bourque SL, Jahroudi N. Aging Is Associated With Organ-Specific Alterations in the Level and Expression Pattern of von Willebrand Factor. Arterioscler Thromb Vasc Biol 2023; 43:2183-2196. [PMID: 37732483 DOI: 10.1161/atvbaha.123.319255] [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: 03/07/2023] [Accepted: 08/31/2023] [Indexed: 09/22/2023]
Abstract
BACKGROUND VWF (von Willebrand factor) is an endothelial-specific procoagulant protein with a major role in thrombosis. Aging is associated with increased circulating levels of VWF, which presents a risk factor for thrombus formation. METHODS Circulating plasma, cellular protein, and mRNA levels of VWF were determined and compared in young and aged mice. Major organs were subjected to immunofluorescence analyses to determine the vascular pattern of VWF expression and the presence of platelet aggregates. An in vitro model of aging, using extended culture time of endothelial cells, was used to explore the mechanism of age-associated increased VWF levels. RESULTS Increased circulating plasma levels of VWF with elevated levels of larger multimers, indicative of VWF functional activity, were observed in aged mice. VWF mRNA and cellular protein levels were significantly increased in the brains, lungs, and livers but not in the kidneys and hearts of aged mice. Higher proportion of small vessels in brains, lungs, and livers of aged mice exhibited VWF expression compared with young, and this was concomitant with increased platelet aggregate formation. Prolonged culture of endothelial cells resulted in increased cell senescence that correlated with increased VWF expression; VWF expression was specifically detected in senescent cultured endothelial cells and abolished in response to p53 knockdown. A significantly higher proportion of VWF expressing endothelial cells in vivo exhibited senescence markers SA-β-Gal (senescence-associated β-galactosidase) and p53 in aged mouse brains compared with that of the young. CONCLUSIONS Aging elicits a heterogenic response in endothelial cells with regard to VWF expression, leading to organ-specific increase in VWF levels and alterations in vascular tree pattern of expression. This is concomitant with increased platelet aggregate formation. The age-associated increase in VWF expression may be modulated through the process of cell senescence, and p53 transcription factor contributes to its regulation.
Collapse
Affiliation(s)
- Parnian Alavi
- Departments of Medicine (P.A., R.Y.A., A.M., N.J.), University of Alberta, Edmonton, Canada
| | - Radya Yousef Abdualla
- Departments of Medicine (P.A., R.Y.A., A.M., N.J.), University of Alberta, Edmonton, Canada
| | - Douglas Brown
- Oncology (D.B., J.L.), University of Alberta, Edmonton, Canada
| | - Anahita Mojiri
- Departments of Medicine (P.A., R.Y.A., A.M., N.J.), University of Alberta, Edmonton, Canada
- Now with Department of Cardiovascular Sciences, Houston Methodist Research Institute, TX (A.M.)
| | | | - John Lewis
- Oncology (D.B., J.L.), University of Alberta, Edmonton, Canada
| | - Stephane L Bourque
- Anesthesiology and Pain Medicine (S.L.B.), University of Alberta, Edmonton, Canada
| | - Nadia Jahroudi
- Departments of Medicine (P.A., R.Y.A., A.M., N.J.), University of Alberta, Edmonton, Canada
| |
Collapse
|
20
|
Casella C, Kiles F, Urquhart C, Michaud DS, Kirwa K, Corlin L. Methylomic, proteomic, and metabolomic correlates of traffic-related air pollution: A systematic review, pathway analysis, and network analysis relating traffic-related air pollution to subclinical and clinical cardiorespiratory outcomes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.30.23296386. [PMID: 37873294 PMCID: PMC10592990 DOI: 10.1101/2023.09.30.23296386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
A growing body of literature has attempted to characterize how traffic-related air pollution (TRAP) affects molecular and subclinical biological processes in ways that could lead to cardiorespiratory disease. To provide a streamlined synthesis of what is known about the multiple mechanisms through which TRAP could lead cardiorespiratory pathology, we conducted a systematic review of the epidemiological literature relating TRAP exposure to methylomic, proteomic, and metabolomic biomarkers in adult populations. Using the 139 papers that met our inclusion criteria, we identified the omic biomarkers significantly associated with short- or long-term TRAP and used these biomarkers to conduct pathway and network analyses. We considered the evidence for TRAP-related associations with biological pathways involving lipid metabolism, cellular energy production, amino acid metabolism, inflammation and immunity, coagulation, endothelial function, and oxidative stress. Our analysis suggests that an integrated multi-omics approach may provide critical new insights into the ways TRAP could lead to adverse clinical outcomes. We advocate for efforts to build a more unified approach for characterizing the dynamic and complex biological processes linking TRAP exposure and subclinical and clinical disease, and highlight contemporary challenges and opportunities associated with such efforts.
Collapse
Affiliation(s)
- Cameron Casella
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Frances Kiles
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Catherine Urquhart
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Dominique S. Michaud
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Kipruto Kirwa
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Laura Corlin
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
- Department of Civil and Environmental Engineering, Tufts University School of Engineering, Medford, MA 02155, USA
| |
Collapse
|
21
|
Płazak W, Drabik L. SARS-CoV-2 infection and SLE: endothelial dysfunction, atherosclerosis, and thrombosis. Clin Rheumatol 2023; 42:2691-2702. [PMID: 36622519 PMCID: PMC9827021 DOI: 10.1007/s10067-022-06497-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 01/10/2023]
Abstract
An increased risk of atherosclerotic and thrombotic complications characterizes connective tissue diseases. Endothelial dysfunction is the basis for the initiation and progression of atherosclerosis and thrombosis. We present systemic lupus erythematosus (SLE) as a model rheumatic disease with endothelial dysfunction and discuss its mechanisms, factors that influence the early onset and rapid progression of atherosclerosis, and the increased risk of thromboembolic events. We focus on established methods to improve endothelium function, including statins, antiplatelet, and antithrombotic therapy. Hypercoagulable and hypofibrinolitic states and a hyperinflammatory response characterize severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Several pathogenic mechanisms are typical for an acute phase of Covid-19 post-Covid syndrome and connective tissue diseases: endothelial dysfunction, elevated antiphospholipid antibody titer, activation of the complement system, and formation of extracellular neutrophil traps (NET). The current review discusses the mechanisms underlying SLE and the COVID-19 in the context of endothelial function, atherosclerosis, and thrombosis (Graphical abstract). Key Points • The pathophysiology of systemic lupus erythematosus (SLE) and Covid-19 shows some similarities, such as endothelial cell activation and dysfunction, the activation of complementary systems, the presence of antiphospholipid antibodies, and the formation of extracellular neutrophil traps. • Autoimmunity in both diseases creates the basis for hyperinflammatory, hypercoagulable, and hypofibrinolitic states and their thromboembolic complications. • This paper presents our perspective on the mechanisms behind the cardiovascular manifestations of SLE and COVID-19, with a particular emphasis on endothelial dysfunction. Covid-19 and systemic lupus erythematosus-potential similarities in pathophysiology. Figures of the panel illustrate the clinical manifestations of endothelial dysfunction, atherosclerosis, and thromboembolism, including coronary artery disease ([A] coronary angiography with left anterior descending artery stenosis and [B] scintigraphy with reduced perfusion in the myocardial apical segments), stroke ([C] carotid angiography, left carotid artery occlusion) and pulmonary embolism ([D]computed tomography with thrombus in the right pulmonary artery).
Collapse
Affiliation(s)
- Wojciech Płazak
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, Jagiellonian University Medical College, Krakow, Poland.
| | - Leszek Drabik
- Department of Cardiac and Vascular Diseases, John Paul II Hospital, Jagiellonian University Medical College, Krakow, Poland
- Department of Pharmacology, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
22
|
Yen CC, Hsu PC, Lin CC, Chen SC, Hsiao CY, Hwang SJ. Effect of far-infrared radiation therapy on von Willebrand factor in patients with chronic kidney disease. Front Med (Lausanne) 2023; 10:1268212. [PMID: 37746066 PMCID: PMC10514495 DOI: 10.3389/fmed.2023.1268212] [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: 07/27/2023] [Accepted: 08/28/2023] [Indexed: 09/26/2023] Open
Abstract
Background Hemostatic abnormality has contributed to vascular access thrombosis in patients with chronic kidney disease (CKD). Previous studies have demonstrated that far-infrared radiation (FIR) therapy can maintain the patency and maturity of arteriovenous fistulas of patients undergoing hemodialysis (HD). However, prolonged access bleeding is observed once FIR is conducted at the end of dialysis. FIR can block the binding of platelet and von Willebrand factor (vWF), a predictor of hemostatic abnormality and vascular access thrombosis. However, clinical studies exploring FIR and vWF are sparse. Methods We recruited 20 HD patients, 21 CKD patients, and 20 controls to examine the alteration of vWF and a disintegrin and metalloproteinase with thrombospondin type 1 repeats 13 (ADAMTS13) following a single 40-min session of FIR therapy. In addition, the alteration of these factors in the HD group was examined following a 40-min FIR session thrice a week for 3 months. Results A decreasing trend in the vWF activity-antigen ratio of participants in all groups following a single FIR session was observed. In addition, the ratio in the HD group was significantly lower following 3 months of FIR therapy. The subgroup analysis revealed a consistent trend and multiple regression analysis showed that participants not taking hydroxymethylglutaryl-coenzyme A reductase inhibitor, diabetes mellitus, and higher hemoglobin levels were the significant factors. The alteration of the vWF activity-antigen ratio correlated moderately to that of ADAMTS13 antigen and activity. Conclusion FIR may alter the ratio of ultra-large vWF multimers through ADAMTS13, contributing to inhibiting platelet-endothelium interactions of CKD patients.
Collapse
Affiliation(s)
- Cheng-Chieh Yen
- Division of Nephrology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi, Taiwan
| | - Po-Chao Hsu
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Ching Lin
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Szu-Chia Chen
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Yen Hsiao
- Division of Nephrology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi, Taiwan
| | - Shang-Jyh Hwang
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli, Taiwan
| |
Collapse
|
23
|
Abstract
Atherosclerosis is the main cause of arterial thrombosis, causing acute occlusive cardiovascular syndromes. Numerous risk prediction models have been developed, which mathematically combine multiple predictors, to estimate the risk of developing cardiovascular events. Current risk models typically do not include information from biomarkers that can potentially improve these existing prediction models especially if they are pathophysiologically relevant. Numerous cardiovascular disease biomarkers have been investigated that have focused on known pathophysiological pathways including those related to cardiac stress, inflammation, matrix remodelling, and endothelial dysfunction. Imaging biomarkers have also been studied that have yielded promising results with a potential higher degree of clinical applicability in detection of atherosclerosis and cardiovascular event prediction. To further improve therapy decision-making and guidance, there is continuing intense research on emerging biologically relevant biomarkers. As the pathogenesis of cardiovascular disease is multifactorial, improvements in discrimination and reclassification in risk prediction models will likely involve multiple biomarkers. This article will provide an overview of the literature on potential blood-based and imaging biomarkers of atherosclerosis studied so far, as well as potential future directions.
Collapse
Affiliation(s)
- Kashan Ali
- From the Division of Molecular & Clinical Medicine, School of Medicine, Ninewells Hospital & Medical School, University of Dundee, Dundee, UK
| | - Chim C Lang
- From the Division of Molecular & Clinical Medicine, School of Medicine, Ninewells Hospital & Medical School, University of Dundee, Dundee, UK
| | - Jeffrey T J Huang
- Biomarker and Drug Analysis Core Facility, Medical Research Institute, Ninewells Hospital & Medical School, University of Dundee, Dundee, UK
| | - Anna-Maria Choy
- From the Division of Molecular & Clinical Medicine, School of Medicine, Ninewells Hospital & Medical School, University of Dundee, Dundee, UK
| |
Collapse
|
24
|
Khan H, Shaikh F, Syed MH, Mamdani M, Saposnik G, Qadura M. Current Biomarkers for Carotid Artery Stenosis: A Comprehensive Review of the Literature. Metabolites 2023; 13:919. [PMID: 37623863 PMCID: PMC10456624 DOI: 10.3390/metabo13080919] [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: 06/30/2023] [Revised: 07/28/2023] [Accepted: 08/03/2023] [Indexed: 08/26/2023] Open
Abstract
Carotid artery stenosis (CAS), an atherosclerotic disease of the carotid artery, is one of the leading causes of transient ischemic attacks (TIA) and cerebrovascular attacks (CVA). The atherogenic process of CAS affects a wide range of physiological processes, such as inflammation, endothelial cell function, smooth muscle cell migration and many more. The current gold-standard test for CAS is Doppler ultrasound; however, there is yet to be determined a strong, clinically validated biomarker in the blood that can diagnose patients with CAS and/or predict adverse outcomes in such patients. In this comprehensive literature review, we evaluated all of the current research on plasma and serum proteins that are current contenders for biomarkers for CAS. In this literature review, 36 proteins found as potential biomarkers for CAS were categorized in to the following nine categories based on protein function: (1) Inflammation and Immunity, (2) Lipid Metabolism, (3) Haemostasis, (4) Cardiovascular Markers, (5) Markers of Kidney Function, (6) Bone Health, (7) Cellular Structure, (8) Growth Factors, and (9) Hormones. This literature review is the most up-to-date and current comprehensive review of research on biomarkers of CAS, and the only review that demonstrated the several pathways that contribute to the initiation and progression of the disease. With this review, future studies can determine if any new markers, or a panel of the proteins explored in this study, may be contenders as diagnostic or prognostic markers for CAS.
Collapse
Affiliation(s)
- Hamzah Khan
- Division of Vascular Surgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (H.K.); (F.S.); (M.H.S.)
- Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada; (M.M.); (G.S.)
| | - Farah Shaikh
- Division of Vascular Surgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (H.K.); (F.S.); (M.H.S.)
- Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada; (M.M.); (G.S.)
| | - Muzammil H. Syed
- Division of Vascular Surgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (H.K.); (F.S.); (M.H.S.)
- Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada; (M.M.); (G.S.)
| | - Muhammad Mamdani
- Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada; (M.M.); (G.S.)
- Temerty Centre for Artificial Intelligence Research and Education in Medicine (T-CAIREM), University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Gustavo Saposnik
- Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON M5B 1W8, Canada; (M.M.); (G.S.)
- Division of Neurology, Department of Medicine, St. Michael’s Hospital, University of Toronto, 55 Queen St E, Toronto, ON M5C 1R6, Canada
| | - Mohammad Qadura
- Division of Vascular Surgery, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (H.K.); (F.S.); (M.H.S.)
- Division of Neurology, Department of Medicine, St. Michael’s Hospital, University of Toronto, 55 Queen St E, Toronto, ON M5C 1R6, Canada
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| |
Collapse
|
25
|
Lilova Z, Hassan F, Riaz M, Ironside J, Ken-Dror G, Han T, Sharma P. Blood group and ischemic stroke, myocardial infarction, and peripheral vascular disease: A meta-analysis of over 145,000 cases and 2,000,000 controls. J Stroke Cerebrovasc Dis 2023; 32:107215. [PMID: 37336185 DOI: 10.1016/j.jstrokecerebrovasdis.2023.107215] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 06/21/2023] Open
Abstract
OBJECTIVE Cardiovascular illnesses have been associated to ABO blood types, specifically through an effect on von Willebrand factor and factor FVIII levels. We conducted a meta-analysis to comprehensively explore the relationship between blood groups and ischemic stroke, myocardial infarction, and peripheral vascular disease. MATERIALS AND METHODS A comprehensive meta-analysis was undertaken to investigate blood groups and ischemic stroke (IS), myocardial infarction (MI) and peripheral vascular disease (PVD). Odds ratios (OR) were used to assess the relationship between blood groups and disease. RevMan v5,4 was used to statistically analyse the results. Risk of bias was assessed using the Newcastle-Ottawa scale. RESULTS A total of 72 studies (18 ischemic stroke, 37 myocardial infarction, 17 peripheral vascular disease) met our search criteria, totalling 145,499 cases and 2,113,736 controls. Mean age ranged between 18 and 90 years. Compared to blood group-O, non-O blood group had an increased association with IS (OR=1.13, 95%Cl: 1.07-1.21, P < 0.001), MI (OR=1.17, 95%Cl: 1.11-1.24, P < 0.001) and PVD (OR=1.15, 95%Cl: 1.04-1.28, P=0.005). Compared to blood group-O, blood group A had a stronger statistically significant association to IS (OR=1.19, P=0.001), MI (OR=1.22, P < 0.001) and PVD (OR=1.15, P=0.03). Blood group-B has the lowest risk associated with MI (OR=1.09, P=0.01). In addition, blood groups AB had a stronger statistically significant association to IS (OR=1.24, P=0.01), and MI (OR=1.20, P < 0.001) compared with the other blood groups. CONCLUSIONS Compared to blood group-O, groups A and AB are strongly associated to ischemic stroke, myocardial infarction, and peripheral vascular disease.
Collapse
Affiliation(s)
- Zornitsa Lilova
- Institute of Cardiovascular Research, Royal Holloway University of London (ICR2UL), London TW20 0EX, UK
| | - Faiza Hassan
- Institute of Cardiovascular Research, Royal Holloway University of London (ICR2UL), London TW20 0EX, UK
| | - Malaika Riaz
- Institute of Cardiovascular Research, Royal Holloway University of London (ICR2UL), London TW20 0EX, UK
| | - Joshua Ironside
- Institute of Cardiovascular Research, Royal Holloway University of London (ICR2UL), London TW20 0EX, UK
| | - Gie Ken-Dror
- Institute of Cardiovascular Research, Royal Holloway University of London (ICR2UL), London TW20 0EX, UK
| | - Thang Han
- Institute of Cardiovascular Research, Royal Holloway University of London (ICR2UL), London TW20 0EX, UK; Department of Endocrinology, Ashford and St Peter's Hospitals NHS Foundation Trust, Surrey, UK
| | - Pankaj Sharma
- Institute of Cardiovascular Research, Royal Holloway University of London (ICR2UL), London TW20 0EX, UK; Department of Neurology, Imperial College Healthcare NHS Trust, UK.
| |
Collapse
|
26
|
Amalia M, Puteri MU, Saputri FC, Sauriasari R, Widyantoro B. Platelet Glycoprotein-Ib (GPIb) May Serve as a Bridge between Type 2 Diabetes Mellitus (T2DM) and Atherosclerosis, Making It a Potential Target for Antiplatelet Agents in T2DM Patients. Life (Basel) 2023; 13:1473. [PMID: 37511848 PMCID: PMC10381765 DOI: 10.3390/life13071473] [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: 05/29/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a persistent metabolic condition that contributes to the development of cardiovascular diseases. Numerous studies have provided evidence that individuals with T2DM are at a greater risk of developing cardiovascular diseases, typically two to four times more likely than those without T2DM, mainly due to an increased risk of atherosclerosis. The rupture of an atherosclerotic plaque leading to pathological thrombosis is commonly recognized as a significant factor in advancing cardiovascular diseases caused by TD2M, with platelets inducing the impact of plaque rupture in established atherosclerosis and predisposing to the primary expansion of atherosclerosis. Studies suggest that individuals with T2DM have platelets that display higher baseline activation and reactivity than those without the condition. The expression enhancement of several platelet receptors is known to regulate platelet activation signaling, including platelet glycoprotein-Ib (GPIb). Furthermore, the high expression of platelet GP1b has been reported to increase the risk of platelet adhesion, platelet-leucocyte interaction, and thrombo-inflammatory pathology. However, the study exploring the role of GP1b in promoting platelet activation-induced cardiovascular diseases in T2DM patients is still limited. Therefore, we summarize the important findings regarding pathophysiological continuity between T2DM, platelet GPIb, and atherosclerosis and highlight the potential therapy targeting GPIb as a novel antiplatelet agent for preventing further cardiovascular incidents in TD2M patients.
Collapse
Affiliation(s)
- Muttia Amalia
- Doctoral Program, Faculty of Pharmacy, Universitas Indonesia, Kampus UI Depok, Depok 16424, Indonesia
| | - Meidi Utami Puteri
- Laboratory of Pharmacology-Toxicology, Faculty of Pharmacy, Universitas Indonesia, Kampus UI Depok, Depok 16424, Indonesia
| | - Fadlina Chany Saputri
- Laboratory of Pharmacology-Toxicology, Faculty of Pharmacy, Universitas Indonesia, Kampus UI Depok, Depok 16424, Indonesia
| | - Rani Sauriasari
- Faculty of Pharmacy, Universitas Indonesia, Kampus UI Depok, Depok 16424, Indonesia
| | - Bambang Widyantoro
- National Cardiovascular Center Harapan Kita, Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Indonesia, Jakarta 11420, Indonesia
| |
Collapse
|
27
|
Jalal MM, Mir R, Hamadi A, Altayar MA, Elfaki I, Barnawi J, Alkayyal AA, Amr M, Hadeel J, Moawadh MS, Alsaedi BSO, Alhelali MH, Yousif A. Association of Genetic and Allelic Variants of Von Willebrand Factor (VWF), Glutathione S-Transferase and Tumor Necrosis Factor Alpha with Ischemic Stroke Susceptibility and Progression in the Saudi Population. Life (Basel) 2023; 13:life13051200. [PMID: 37240845 DOI: 10.3390/life13051200] [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: 03/09/2023] [Revised: 04/25/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Stroke is a key cerebrovascular disease and important cause of death and disability worldwide, including in the kingdom of Saudi Arabia (KSA). It has a large economic burden and serious socioeconomic impacts on patients, their families and the community. The incidence of ischemic stroke is probably increased by the interaction of GSTT1 and GSTM1 null genotypes with high blood pressure, diabetes and cigarette smoking. The roles of VWF, GSTs and TNF-alpha gene variations in the induction of stroke are still uncertain and require further examination. In the current study, we studied the associations of SNPs in the genes VWF, GSTs and TNF-alpha with stroke in the Saudi population. Genotyping was performed using the ARMS -PCR for TNF-alpha, AS-PCR for VWF and multiplex PCR for GSTs. The study included 210 study subjects: 100 stroke cases and 110 healthy controls. We obtained significant distributions of VWF rs61748511 T > C, TNF-alpha rs1800629 G > A and GST rs4025935 and rs71748309 genotypes between stroke cases and the healthy controls (p < 0.05). The results also indicated that the TNF-alpha A allele was associated with risk of stroke with odd ratio (OR) = 2.22 and risk ratio = RR 2.47, p < 0.05. Similarly, the VWF-TC genotype and C allele were strongly linked with stroke with OR = 8.12 and RR 4.7, p < 0.05. In addition, GSTT1 and GSTT1 null genotype was strongly associated with stroke predisposition with OR = 8.30 and RR = 2.25, p < 0.0001. We conclude that there is a possible strong association between the VWF-T > C, TNF-alpha G > A, GSTT1 gene variants and ischemic stroke susceptibility in the Saudi population. However, future well-designed and large-scale case-control studies on protein-protein interactions and protein functional studies are required to verify these findings and examine the effects of these SNPs on these proteins.
Collapse
Affiliation(s)
- Mohammed M Jalal
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
- Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Rashid Mir
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
- Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Abdullah Hamadi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
- Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Malik A Altayar
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
- Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Imadeldin Elfaki
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Jameel Barnawi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
- Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Almohanad A Alkayyal
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Mouminah Amr
- Neuroscience Center, King Abdullah Medical Complex, Jeddah 23816, Saudi Arabia
| | - Jabali Hadeel
- Department of Radiology, King Abdullah Medical Complex, Jeddah 23816, Saudi Arabia
| | - Mamdoh S Moawadh
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Basim S O Alsaedi
- Department of Statistics, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Marwan H Alhelali
- Department of Statistics, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Aadil Yousif
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| |
Collapse
|
28
|
Mitrofanova LB, Makarov IA, Gorshkov AN, Runov AL, Vonsky MS, Pisareva MM, Komissarov AB, Makarova TA, Li Q, Karonova TL, Konradi AO, Shlaykhto EV. Comparative Study of the Myocardium of Patients from Four COVID-19 Waves. Diagnostics (Basel) 2023; 13:diagnostics13091645. [PMID: 37175037 PMCID: PMC10178873 DOI: 10.3390/diagnostics13091645] [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: 03/17/2023] [Revised: 04/27/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND Few studies have compared COVID-19 patients from different waves. This study aims to conduct a clinical and morphological analysis of patients who died from COVID-19 during four waves. METHODS The study involved 276 patients who died from COVID-19 during four waves, including 77 patients in the first wave, 119 patients in the second wave, and 78 patients in the third wave. We performed a histological examination of myocardium samples from autopsies and additionally analyzed the samples by PCR. We conducted immunohistochemistry of the myocardium for 21 samples using antibodies against CD3, CD45, CD8, CD68, CD34, Ang1, VWF, VEGF, HLA-DR, MHC1, C1q, enteroviral VP1, and SARS-CoV-2 spike protein. We also did immunofluorescent staining of three myocardial specimens using VP1/SARS-CoV-2 antibody cocktails. Further, we ran RT-ddPCR analysis for 14 RNA samples extracted from paraffin-embedded myocardium. Electron microscopic studies of the myocardium were also performed for two samples from the fourth wave. RESULTS Among the 276 cases, active myocarditis was diagnosed in 5% (15/276). Of these cases, 86% of samples expressed VP1, and individual cells contained SARS-CoV-2 spike protein in 22%. Immunofluorescence confirmed the co-localization of VP1 and SARS-CoV-2 spike proteins. ddPCR did not confidently detect SARS-CoV-2 RNA in the myocardium in any myocarditis cases. However, the myocardium sample from wave IV detected a sub-threshold signal of SARS-CoV-2 by qPCR, but myocarditis in this patient was not confirmed. Electron microscopy showed several single particles similar to SARS-CoV-2 virions on the surface of the endothelium of myocardial vessels. A comparison of the cardiovascular complication incidence between three waves revealed that the incidence of hemorrhage (48 vs. 24 vs. 17%), myocardial necrosis (18 vs. 11 vs. 4%), blood clots in the intramural arteries (12 vs. 7 vs. 0%), and myocarditis (19 vs. 1 vs. 6%) decreased over time, and CD8-T-killers appeared. Immunohistochemistry confirmed the presence of endotheliitis in all 21 studied cases. CONCLUSIONS This study compared myocardial damage in patients who died during three COVID-19 waves and showed a decrease in the incidence of endotheliitis complications (thrombosis, hemorrhage, necrosis) and myocarditis over time. However, the connection between myocarditis and SARS-CoV-2 infection remains unproven.
Collapse
Affiliation(s)
| | | | - Andrey Nikolaevich Gorshkov
- Almazov National Medical Research Centre, St. Petersburg 197341, Russia
- Smorodintsev Research Institute of Influenza, St. Petersburg 197376, Russia
| | - Andrey Leonidovich Runov
- Almazov National Medical Research Centre, St. Petersburg 197341, Russia
- D.I. Mendeleyev Institute for Metrology, St. Petersburg 190005, Russia
| | - Maxim Sergeevich Vonsky
- Almazov National Medical Research Centre, St. Petersburg 197341, Russia
- D.I. Mendeleyev Institute for Metrology, St. Petersburg 190005, Russia
| | | | | | | | - Qingli Li
- East China Normal University, Shanghai 200241, China
| | | | | | | |
Collapse
|
29
|
Rehman M, Chaudhary R, Rajput S, Agarwal V, Kaushik AS, Srivastava S, Srivastava S, Singh R, Aziz I, Singh S, Mishra V. Butein Ameliorates Chronic Stress Induced Atherosclerosis via Targeting Anti-inflammatory, Anti-fibrotic and BDNF Pathways. Physiol Behav 2023; 267:114207. [PMID: 37100219 DOI: 10.1016/j.physbeh.2023.114207] [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: 01/31/2023] [Revised: 03/31/2023] [Accepted: 04/23/2023] [Indexed: 04/28/2023]
Abstract
Chronic stress is a major risk factor for various diseases, including cardiovascular diseases (CVDs). Chronic stress enhances the release of pro-inflammatory cytokines like IL-1β, IL-6, and TNF-α, making individuals susceptible to atherosclerosis which is dominant cause for CVDs. In present study, we validated a mouse model of chronic unpredictable stress (CUS), and assessed the characteristic features of atherosclerosis in thoracic aortas of CUS mice. The CUS procedure consisted of exposing groups of mice to random stressors daily for 10-weeks. The stress response was verified by presence of depressive-like behaviors and increased serum corticosterone in mice which was determined by battery of behavioural tests (SPT, EPMT, NSFT) and ELISA, respectively. Atherosclerosis parameters in CUS mice were evaluated by lipid indices estimation followed by histological assessment of plaque deposition and fibrosis in thoracic aorta. Further, we assessed the efficacy of a polyphenol, i.e. Butein in conferring protection against chronic stress-induced atherosclerosis and the possible mechanism of action. Butein (20mg/kg x 28 days, alternatively, i.p.) was administered to CUS mice after 6-weeks of CUS exposure till the end of the protocol. Butein treatment decreased peripheral IL-1β and enhanced peripheral as well as central BDNF levels. Histological assessment revealed decreased macrophage expression and reduced fibrosis in thoracic aorta of Butein treated mice. Further, treatment with Butein lowered lipid indices in CUS mice. Our findings thus, suggest that 10-weeks of CUS induce characteristic features of atherosclerosis in mice and Butein can offer protection in CUS-induced atherosclerosis through multiple mechanisms including anti-inflammatory, antifibrotic and anti-adipogenic actions.
Collapse
Affiliation(s)
- Mujeeba Rehman
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025(.) India
| | - Rishabh Chaudhary
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025(.) India
| | - Sonu Rajput
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025(.) India
| | - Vipul Agarwal
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025(.) India
| | - Arjun Singh Kaushik
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025(.) India
| | - Siddhi Srivastava
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025(.) India
| | - Sukriti Srivastava
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025(.) India
| | - Rohit Singh
- Centre for Animal Disease Research and Diagnosis, ICAR-Indian Veterinary Research Institute, Izzatnagar, Bareilly 243122, Uttar Pradesh, India
| | - Irfan Aziz
- Integral University, Kursi road, Lucknow, Uttar Pradesh 226026, India
| | - Sanjay Singh
- Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025, India.
| | - Vikas Mishra
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow, Uttar Pradesh 226025(.) India.
| |
Collapse
|
30
|
Kennedy KG, Mio M, Goldstein BI, Brambilla P, Delvecchio G. Systematic review and meta-analysis of retinal microvascular caliber in bipolar disorder, major depressive disorder, and schizophrenia. J Affect Disord 2023; 331:342-351. [PMID: 36958491 DOI: 10.1016/j.jad.2023.03.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/06/2023] [Accepted: 03/09/2023] [Indexed: 03/25/2023]
Abstract
BACKGROUND Individuals with a severe mental illness (SMI), such as bipolar disorder (BD), major depressive disorder (MDD), and schizophrenia (SZ), have increased rates of cardiovascular and cerebrovascular disease. Interestingly, it has been reported that retinal microvessels, a proxy cerebrovascular measure, non-invasively assessed via retinal imaging, predict future cardiovascular disease, with some studies also showing anomalous retinal microvascular caliber in SMI. Therefore, this review and meta-analysis evaluated whether retinal microvascular caliber differs between individuals with SMI vs controls and summarized current findings. METHODS A systematic literature search for retinal microvascular caliber and SMI was conducted in Embase and MEDLINE. Studies needed to be published in English before 2022 December 1st and examine retinal microvascular caliber in individuals diagnosed with a SMI. Finally, a meta-analysis of arteriolar and venular caliber in SMI case-controlled studies was also conducted. RESULTS The search yielded 65 unique articles, 11 were included in the review and 6 in the meta-analysis. The meta-analysis found that the SMI group had significantly wider venules than controls (SMD = -0.53; 95 % CI = 0.24, 0.81; p = 0.0004) but not arterioles (SMD = 0.07; 95 % CI = -0.29, 0.44; p = 0.70). Additionally, the systematic review found that poorer retinal microvascular health is associated with greater illness severity. LIMITATIONS Large heterogeneity of findings and small sample size. CONCLUSION This systematic review and meta-analysis found that SMI, specifically SZ, is associated with wider retinal venules. Retinal imaging, a fast, cost-effective, and non-invasive assay of cerebrovascular health, may provide insight into the pathophysiological processes of SMI. However, future longitudinal studies investigating these findings are warranted.
Collapse
Affiliation(s)
- Kody G Kennedy
- Centre for Youth Bipolar Disorder, Centre for Addictions and Mental Health, Toronto, Canada; Department of Pharmacology, University of Toronto, Toronto, Canada
| | - Megan Mio
- Centre for Youth Bipolar Disorder, Centre for Addictions and Mental Health, Toronto, Canada; Department of Pharmacology, University of Toronto, Toronto, Canada
| | - Benjamin I Goldstein
- Centre for Youth Bipolar Disorder, Centre for Addictions and Mental Health, Toronto, Canada; Department of Pharmacology, University of Toronto, Toronto, Canada
| | - Paolo Brambilla
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Giuseppe Delvecchio
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| |
Collapse
|
31
|
Stritt S, Nurden P, Nurden AT, Schved JF, Bordet JC, Roux M, Alessi MC, Trégouët DA, Mäkinen T, Giansily-Blaizot M. APOLD1 loss causes endothelial dysfunction involving cell junctions, cytoskeletal architecture, and Weibel-Palade bodies, while disrupting hemostasis. Haematologica 2023; 108:772-784. [PMID: 35638551 PMCID: PMC9973481 DOI: 10.3324/haematol.2022.280816] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Indexed: 11/09/2022] Open
Abstract
Vascular homeostasis is impaired in various diseases thereby contributing to the progression of their underlying pathologies. The endothelial immediate early gene Apolipoprotein L domain-containing 1 (APOLD1) helps to regulate endothelial function. However, its precise role in endothelial cell biology remains unclear. We have localized APOLD1 to endothelial cell contacts and to Weibel-Palade bodies (WPB) where it associates with von Willebrand factor (VWF) tubules. Silencing of APOLD1 in primary human endothelial cells disrupted the cell junction-cytoskeletal interface, thereby altering endothelial permeability accompanied by spontaneous release of WPB contents. This resulted in an increased presence of WPB cargoes, notably VWF and angiopoietin-2 in the extracellular medium. Autophagy flux, previously recognized as an essential mechanism for the regulated release of WPB, was impaired in the absence of APOLD1. In addition, we report APOLD1 as a candidate gene for a novel inherited bleeding disorder across three generations of a large family in which an atypical bleeding diathesis was associated with episodic impaired microcirculation. A dominant heterozygous nonsense APOLD1:p.R49* variant segregated to affected family members. Compromised vascular integrity resulting from an excess of plasma angiopoietin-2, and locally impaired availability of VWF may explain the unusual clinical profile of APOLD1:p.R49* patients. In summary, our findings identify APOLD1 as an important regulator of vascular homeostasis and raise the need to consider testing of endothelial cell function in patients with inherited bleeding disorders without apparent platelet or coagulation defects.
Collapse
Affiliation(s)
- Simon Stritt
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala
| | - Paquita Nurden
- Institut de Rythmologie et de Modélisation Cardiaque, Hôpital Xavier Arnozan, Pessac, France.
| | - Alan T Nurden
- Institut de Rythmologie et de Modélisation Cardiaque, Hôpital Xavier Arnozan, Pessac, France
| | - Jean-François Schved
- Department of Biological Hematology, CHU Montpellier, Université de Montpellier, Montpellier
| | - Jean-Claude Bordet
- Hematology, Hospices civils de Lyon, Bron biology center and Hemostasis- Thrombosis, Lyon-1 University, Lyon
| | | | | | - David-Alexandre Trégouët
- Laboratory of Excellence GENMED (Medical Genomics), Paris; University of Bordeaux, INSERM, Bordeaux Population Health Research Center, U1219, Bordeaux
| | - Taija Mäkinen
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Muriel Giansily-Blaizot
- Department of Biological Hematology, CHU Montpellier, Université de Montpellier, Montpellier
| |
Collapse
|
32
|
Galectin-3 and Blood Group: Binding Properties, Effects on Plasma Levels, and Consequences for Prognostic Performance. Int J Mol Sci 2023; 24:ijms24054415. [PMID: 36901846 PMCID: PMC10002292 DOI: 10.3390/ijms24054415] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/08/2023] [Accepted: 02/14/2023] [Indexed: 02/25/2023] Open
Abstract
Previous studies have reported an association between ABO type blood group and cardiovascular (CV) events and outcomes. The precise mechanisms underpinning this striking observation remain unknown, although differences in von Willebrand factor (VWF) plasma levels have been proposed as an explanation. Recently, galectin-3 was identified as an endogenous ligand of VWF and red blood cells (RBCs) and, therefore, we aimed to explore the role of galectin-3 in different blood groups. Two in vitro assays were used to assess the binding capacity of galectin-3 to RBCs and VWF in different blood groups. Additionally, plasma levels of galectin-3 were measured in different blood groups in the Ludwigshafen Risk and Cardiovascular Health (LURIC) study (2571 patients hospitalized for coronary angiography) and validated in a community-based cohort of the Prevention of Renal and Vascular End-stage Disease (PREVEND) study (3552 participants). To determine the prognostic value of galectin-3 in different blood groups, logistic regression and cox regression models were used with all-cause mortality as the primary outcome. First, we demonstrated that galectin-3 has a higher binding capacity for RBCs and VWF in non-O blood groups, compared to blood group O. Additionally, LURIC patients with non-O blood groups had substantially lower plasma levels of galectin-3 (15.0, 14.9, and 14.0 μg/L in blood groups A, B, and AB, respectively, compared to 17.1 μg/L in blood group O, p < 0.0001). Finally, the independent prognostic value of galectin-3 for all-cause mortality showed a non-significant trend towards higher mortality in non-O blood groups. Although plasma galectin-3 levels are lower in non-O blood groups, the prognostic value of galectin-3 is also present in subjects with a non-O blood group. We conclude that physical interaction between galectin-3 and blood group epitopes may modulate galectin-3, which may affect its performance as a biomarker and its biological activity.
Collapse
|
33
|
Wang K, Wang W, Lei L, Lan Y, Liu Q, Ren L, Wu S. Association between short-term exposure to ambient air pollution and biomarkers of coagulation: A systematic review and meta-analysis. ENVIRONMENTAL RESEARCH 2022; 215:114210. [PMID: 36030918 DOI: 10.1016/j.envres.2022.114210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 06/15/2023]
Abstract
Ambient air pollution is one of the major global risk factors for cardiovascular health, and coagulation changes have been proposed to mediate this risk. Plasminogen activator inhibitor-1 (PAI-1), von Willebrand factor (vWF), soluble P-selectin (sP-selectin) and tissue plasminogen activator (t-PA) are major coagulation biomarkers. However, there has been no systematic meta-analysis to summarize associations of ambient air pollution with these coagulation biomarkers. To assess the overall associations between ambient particulate matter (PM2.5, PM10), ozone (O3), nitrogen dioxide (NO2), carbon monoxide (CO) and major coagulation biomarkers including PAI-1, vWF, sP-selectin and t-PA based on the existing epidemiological research. We performed a systematic literature search of publications reporting the associations of ambient air pollutants (PM2.5, PM10, O3, NO2, and CO) with coagulation biomarkers (PAI-1, vWF, sP-selectin and t-PA) in PubMed, Web of Science, EMBASE, and Scopus databases as of April 5, 2022. Then, we performed a random-effect meta-analysis, which included 27 articles, and then identified the potential sources of heterogeneity. The pooled percent changes of coagulation biomarkers per 10 μg/m3 increase in short-term exposure to ambient PM2.5 were 2.43% (95% CI: 0.59%, 4.29%) in PAI-1, 1.08% (95% CI: 0.21%, 1.96%) in vWF and 1.14% (95% CI: 0.59%, 1.68%) in sP-selectin, respectively. We also found significant associations of short-term exposure to ambient O3 with PAI-1 (1.62%, 95% CI: 0.01%, 3.25%), sP-selectin (9.59%, 95% CI:2.78%, 16.86%) and t-PA (0.45%, 95% CI: 0.02%, 0.88%), respectively. Short-term exposures to ambient PM10, NO2 and CO were not significantly associated with changes in coagulation biomarkers. In conclusion, short-term exposures to PM2.5 and O3 are associated with significant increases in coagulation biomarkers, suggesting an activated coagulation state upon air pollution exposure.
Collapse
Affiliation(s)
- Kai Wang
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China; Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, Shaanxi, China; Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, China
| | - Wanzhou Wang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, China
| | - Lei Lei
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China; Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, Shaanxi, China; Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, China
| | - Yang Lan
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China; Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, Shaanxi, China; Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, China
| | - Qisijing Liu
- Research Institute of Public Health, School of Medicine, Nankai University, Tianjin, China
| | - Lihua Ren
- School of Nursing, Peking University, Beijing, China
| | - Shaowei Wu
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China; Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, Shaanxi, China; Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, China.
| |
Collapse
|
34
|
Qi X, Hatami S, Bozso S, Buchko M, Forgie KA, Olafson C, Khan M, Himmat S, Wang X, Nobes DS, Acker JP, Nagendran J, Freed DH. The evaluation of constant coronary artery flow versus constant coronary perfusion pressure during normothermic ex situ heart perfusion. J Heart Lung Transplant 2022; 41:1738-1750. [PMID: 36137869 DOI: 10.1016/j.healun.2022.08.009] [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: 09/04/2021] [Revised: 08/10/2022] [Accepted: 08/10/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Evidence suggests that hearts that are perfused under ex-situ conditions lose normal coronary vasomotor tone and experience contractile failure over a few hours. We aimed to evaluate the effect of different coronary perfusion strategies during ex situ heart perfusion on cardiac function and coronary vascular tone. METHODS Porcine hearts (n = 6 each group) were perfused in working mode for 6 hours with either constant aortic diastolic pressure (40 mmHg) or constant coronary flow rate (500 mL/min). Functional and metabolic parameters, cytokine profiles, cardiac and vascular injury, coronary artery function and oxidative stress were compared between groups. RESULTS Constant coronary flow perfusion demonstrated better functional preservation and less edema formation (Cardiac index: flow control = 8.33 vs pressure control = 6.46 mL·min-1·g-1, p = 0.016; edema formation: 7.92% vs 19.80%, p < 0.0001). Pro-inflammatory cytokines, platelet activation as well as endothelial activation were lower in the flow control group. Similarly, less cardiac and endothelial injury was observed in the constant coronary flow group. Evaluation of coronary artery function showed there was loss of coronary autoregulation in both groups. Oxidative stress was induced in the coronary arteries and was relatively lower in the flow control group. CONCLUSIONS A strategy of controlled coronary flow during ex situ heart perfusion provides superior functional preservation and less edema formation, together with less myocardial damage, leukocyte, platelet, endothelial activation, and oxidative stress. There was loss of coronary autoregulation and decrease of coronary vascular resistance during ESHP irrespective of coronary flow control strategy. Inflammation and oxidative stress state in the coronary vasculature may play a role.
Collapse
Affiliation(s)
- Xiao Qi
- Departments of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Sanaz Hatami
- Departments of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Sabin Bozso
- Departments of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Max Buchko
- Departments of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Keir A Forgie
- Departments of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Carly Olafson
- Centre for Innovation, Canadian Blood Services, Edmonton, AB, Canada
| | - Mubashir Khan
- Departments of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Sayed Himmat
- Departments of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Xiuhua Wang
- Departments of Surgery, University of Alberta, Edmonton, AB, Canada
| | - David S Nobes
- Department of Mechanical Engineering, Faculty of Engineering, University of Alberta, AB, Canada
| | - Jason P Acker
- Department of Laboratory Medicine and Pathology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada; Centre for Innovation, Canadian Blood Services, Edmonton, AB, Canada
| | - Jayan Nagendran
- Departments of Surgery, University of Alberta, Edmonton, AB, Canada; Alberta Transplant Institute Edmonton, AB, Canada; Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada
| | - Darren H Freed
- Departments of Surgery, University of Alberta, Edmonton, AB, Canada; Physiology, University of Alberta, Edmonton, AB, Canada; Biomedical Engineering, University of Alberta, Edmonton, AB, Canada; Alberta Transplant Institute Edmonton, AB, Canada; Canadian Donation and Transplantation Research Program, Edmonton, AB, Canada.
| |
Collapse
|
35
|
van Paridon PCS, Panova‐Noeva M, van Oerle R, Schulz A, Prochaska JH, Arnold N, Schmidtmann I, Beutel M, Pfeiffer N, Münzel T, Lackner KJ, ten Cate H, Wild PS, Spronk HMH. Lower levels of vWF are associated with lower risk of cardiovascular disease. Res Pract Thromb Haemost 2022; 6:e12797. [PMID: 36381288 PMCID: PMC9637545 DOI: 10.1002/rth2.12797] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 07/23/2022] [Accepted: 07/29/2022] [Indexed: 11/09/2022] Open
Abstract
Objective The current study was undertaken to prospectively explore whether having low levels of von Willebrand factor (vWF) antigen and vWF activity reduce the risk for cardiovascular disease and death. Methods VWF antigen and vWF activity were measured by enzyme-linked immunosorbent assay and an immunological-based assay, respectively, in a subsample of 4857 individuals aged between 35 and 74 years old, enrolled between April 2007 and October 2008 in the population-based Gutenberg Health Study. VWF antigen and activity below the 20th percentile was set as a measure of "low vWF." Adjusted robust Poisson regression models were used to analyze the relation between low vWF and the incidence of cardiovascular disease (CVD). Consequent adjusted cox regression models as well as cumulative incidence plots were calculated to explore the relation between all-cause and cardiovascular mortality and low vWF. Results VWF activity levels <20th percentile (i.e., <76.2%) were associated with a decreased relative risk for CVD (RR: 0.59, 95% CI: 0.37-0.95), despite adjusting for age and sex. After adjusting for levels of F-VIII, the association persisted (RR: 0.60, 95% CI: 0.36-0.99). The cumulative incidence plots demonstrated that vWF antigen <20th percentile significantly correlated with decreased cardiovascular mortality. VWF antigen<20th percentile (i.e., <83%) was significantly associated with lower risk of all-cause mortality, despite adjusting for clinical factors (RR: 61, 95% CI: 0.41-0.91). Conclusion The study demonstrated that having low vWF activity levels were associated with a lower risk for CVD. Additionally, it revealed a decreased risk of cardiovascular and all-cause mortality in individuals with low levels of vWF antigen, shining new light on vWF as a potential target for novel therapies.
Collapse
Affiliation(s)
- Pauline C. S. van Paridon
- Laboratory for Clinical Thrombosis and Hemostasis, Department of Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM)Maastricht University Medical CenterMaastrichtThe Netherlands
- Center for Thrombosis and Hemostasis (CTH)University Medical Center of the Johannes Gutenberg‐University MainzMainzGermany
| | - Marina Panova‐Noeva
- Center for Thrombosis and Hemostasis (CTH)University Medical Center of the Johannes Gutenberg‐University MainzMainzGermany
- DZHK (German Center for Cardiovascular Research)Partner Site RhineMainMainzGermany
| | - Rene van Oerle
- Laboratory for Clinical Thrombosis and Hemostasis, Department of Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM)Maastricht University Medical CenterMaastrichtThe Netherlands
| | - Andreas Schulz
- Preventive Cardiology and Preventive Medicine, Center for CardiologyUniversity Medical Center of the Johannes Gutenberg‐University MainzMainzGermany
| | - Jürgen H. Prochaska
- Center for Thrombosis and Hemostasis (CTH)University Medical Center of the Johannes Gutenberg‐University MainzMainzGermany
- DZHK (German Center for Cardiovascular Research)Partner Site RhineMainMainzGermany
- Preventive Cardiology and Preventive Medicine, Center for CardiologyUniversity Medical Center of the Johannes Gutenberg‐University MainzMainzGermany
| | - Natalie Arnold
- Preventive Cardiology and Preventive Medicine, Center for CardiologyUniversity Medical Center of the Johannes Gutenberg‐University MainzMainzGermany
| | - Irene Schmidtmann
- Institute of Medical Biostatistics, Epidemiology and InformaticsUniversity Medical Center of the Johannes Gutenberg‐University MainzMainzGermany
| | - Manfred Beutel
- Department of Psychosomatic Medicine and PsychotherapyUniversity Medical Center of the Johannes Gutenberg‐University MainzMainzGermany
| | - Norbert Pfeiffer
- Department of OphthalmologyUniversity Medical Center of the Johannes Gutenberg‐University MainzMainzGermany
| | - Thomas Münzel
- DZHK (German Center for Cardiovascular Research)Partner Site RhineMainMainzGermany
- Center for Cardiology IUniversity Medical Center of the Johannes Gutenberg‐University MainzMainzGermany
| | - Karl J. Lackner
- DZHK (German Center for Cardiovascular Research)Partner Site RhineMainMainzGermany
- Institute for Clinical Chemistry and Laboratory MedicineUniversity Medical Center of the Johannes Gutenberg‐University MainzMainzGermany
| | - Hugo ten Cate
- Laboratory for Clinical Thrombosis and Hemostasis, Department of Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM)Maastricht University Medical CenterMaastrichtThe Netherlands
- Center for Thrombosis and Hemostasis (CTH)University Medical Center of the Johannes Gutenberg‐University MainzMainzGermany
| | - Philipp S. Wild
- Center for Thrombosis and Hemostasis (CTH)University Medical Center of the Johannes Gutenberg‐University MainzMainzGermany
- DZHK (German Center for Cardiovascular Research)Partner Site RhineMainMainzGermany
- Preventive Cardiology and Preventive Medicine, Center for CardiologyUniversity Medical Center of the Johannes Gutenberg‐University MainzMainzGermany
| | - Henri M. H. Spronk
- Laboratory for Clinical Thrombosis and Hemostasis, Department of Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM)Maastricht University Medical CenterMaastrichtThe Netherlands
| |
Collapse
|
36
|
Hirai H, Yamashita M, Matsumoto M, Nishiyama T, Wada D, Okabe N, Mizusawa Y, Jimura H, Ueda T, Ogata N. Alteration of plasma von Willebrand factor in the treatment of retinal vein occlusion with cystoid macular edema. PLoS One 2022; 17:e0264809. [PMID: 36137144 PMCID: PMC9499207 DOI: 10.1371/journal.pone.0264809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 09/08/2022] [Indexed: 11/18/2022] Open
Abstract
Retinal vein occlusion (RVO) is a major retinal disease caused by venous thrombosis. Although several studies have proposed an association between venous thrombosis and von Willebrand factor (VWF), the association between RVO and VWF remains unclear. We aimed to investigate the association between RVO and VWF and the alteration of VWF levels under anti-vascular endothelial growth factor (VEGF) treatment. We enrolled 55 patients with RVO involved cystoid macular edema. They received intravitreal injection of anti-VEGF drugs, either ranibizumab or aflibercept. We examined the clinical data and measured plasma VWF antigen and a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13 (ADAMTS13) activity to identify variabilities during treatment. At baseline, there was no significant difference between the RVO group and age-matched controls in both VWF antigen and ADAMTS13 activity levels, but ADAMTS13 activity was significantly lower in central RVO than in branch RVO (P = 0.015). In branch RVO, VWF antigen was negatively correlated with central choroidal thickness (r = −0.51, P < 0.001). In branch RVO after anti-VEGF treatment, VWF antigen levels decreased significantly from 134% at baseline to 109% at 1 day (P = 0.002) and 107% at 1 month (P = 0.030) after treatment. In contrast, ADAMTS13 activity showed no significant difference during this period. In branch RVO at 1 month after treatment, VWF antigen was negatively correlated with central choroidal thickness (r = −0.47, P = 0.001). Our findings suggest an association between VWF and central choroidal thickness in patients with branch RVO, thus the measurement of VWF may be useful for evaluating disease activity and prognosis.
Collapse
Affiliation(s)
- Hiromasa Hirai
- Department of Ophthalmology, Nara Medical University, Kashihara, Japan
| | | | - Masanori Matsumoto
- Department of Blood Transfusion Medicine, Nara Medical University, Kashihara, Japan
| | | | - Daishi Wada
- Department of Ophthalmology, Nara Medical University, Kashihara, Japan
| | - Naoko Okabe
- Department of Ophthalmology, Nara Medical University, Kashihara, Japan
| | - Yutaro Mizusawa
- Department of Ophthalmology, Nara Medical University, Kashihara, Japan
| | - Hironobu Jimura
- Department of Ophthalmology, Nara Medical University, Kashihara, Japan
| | - Tetsuo Ueda
- Department of Ophthalmology, Nara Medical University, Kashihara, Japan
| | - Nahoko Ogata
- Department of Ophthalmology, Nara Medical University, Kashihara, Japan
- * E-mail:
| |
Collapse
|
37
|
Plasma Angiotensin Converting Enzyme 2 (ACE2) Activity in Healthy Controls and Patients with Cardiovascular Risk Factors and/or Disease. J Pers Med 2022; 12:jpm12091495. [PMID: 36143280 PMCID: PMC9501250 DOI: 10.3390/jpm12091495] [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: 08/10/2022] [Revised: 09/05/2022] [Accepted: 09/08/2022] [Indexed: 11/18/2022] Open
Abstract
Angiotensin converting enzyme 2 (ACE2) is an endogenous negative regulator of the renin-angiotensin system, a key factor in the development of cardiovascular disease (CVD). ACE2 is also used by SARS-CoV-2 for host cell entry. Given that COVID-19 is associated with hypercoagulability, it is timely to explore the potential relationship between plasma ACE2 activity and the coagulation profile. In this cross-sectional study, ACE2 activity and global coagulation assays (GCA) including thromboelastography, thrombin, and fibrin generation were measured in adult healthy controls (n = 123; mean age 41 ± 17 years; 35% male) and in patients with cardiovascular risk factors and/or disease (n = 258; mean age 65 ± 14 years; 55% male). ACE2 activity was significantly lower in controls compared to patients with cardiovascular risk factors and/or disease (median 0.10 (0.02, 3.33) vs. 5.99 (1.95, 10.37) pmol/mL/min, p < 0.001). Of the healthy controls, 48% had undetectable ACE2 activity. Controls with detectable ACE2 had lower maximum amplitude (p < 0.001). In patients with cardiovascular risk factors and/or disease, those in the 3rd tertile were older and male (p = 0.002), with a higher Framingham grade and increased number of cardiovascular risk factors (p < 0.001). In conclusion, plasma ACE2 activity is undetectable to very low in young healthy controls with minimal clinically relevant associations to GCA. Patients with cardiovascular risk factors and/or disease have increased plasma ACE2 activity, suggesting that it may be an important biomarker of endothelial dysfunction and atherosclerosis.
Collapse
|
38
|
Illési Á, Debreceni IB, Fejes Z, Nagy B, Hodosi K, Kappelmayer J, Csanádi Z, Szük TI. Effect of invasive therapeutic coronary interventions on endothelial cell activation and thrombin generation in patients with chronic total coronary occlusion. Thromb Res 2022; 217:64-72. [PMID: 35908382 DOI: 10.1016/j.thromres.2022.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 07/06/2022] [Accepted: 07/18/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Percutaneous coronary intervention (PCI) is commonly used treatment for chronic total occlusion (CTO). PCI can be performed in two different ways using wire escalation (WE) or subintimal dissection and reentry (DR) technique. During both procedures patients are treated with anticoagulants, however a substantial activation of coagulation cascade is expected, which may affect clinical outcome. OBJECTIVES Our aim was to compare the impact of WE and DR techniques regarding endothelial cell activation and thrombin formation. METHODS Fifty patients after CTO-PCI were enrolled into this study. Blood samples were obtained before PCI, at 48 h and 3-6 months after the intervention to measure soluble endothelium-specific markers and to investigate thrombin generation. RESULTS Twenty-nine patients were treated with WE, 21 received DR. In the DR group, soluble VCAM-1 (vascular cell adhesion molecule-1) and ICAM-1 (intercellular cell adhesion molecule-1) concentrations were gradually elevated and remained significantly increased at 3-6 months (p = 0.006 and p = 0.037, respectively) compared to pre-PCI. Furthermore, significant decrease in lagtime (p = 0.004) and time to peak (p = 0.002) with a substantial increment in peak thrombin (p = 0.001) were observed in these patients. In contrast, no significant alteration was found in the WE cohort. Clinical complications (myocardial infarction, stroke, thrombosis, revascularization) did not occur in the first 9 months of follow-up period in either group. CONCLUSION Although DR intervention induces more thrombin generation with a larger degree of endothelium activation compared to WE, this technique does not cause more clinical complications.
Collapse
Affiliation(s)
- Ádám Illési
- Department of Cardiology and Cardiac Surgery, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., H-4032 Debrecen, Hungary; Doctoral School of Kálmán Laki, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., H-4032 Debrecen, Hungary
| | - Ildikó Beke Debreceni
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., H-4032 Debrecen, Hungary
| | - Zsolt Fejes
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., H-4032 Debrecen, Hungary
| | - Béla Nagy
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., H-4032 Debrecen, Hungary
| | - Katalin Hodosi
- Institute of Internal Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., H-4032 Debrecen, Hungary
| | - János Kappelmayer
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., H-4032 Debrecen, Hungary
| | - Zoltán Csanádi
- Department of Cardiology and Cardiac Surgery, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., H-4032 Debrecen, Hungary; Doctoral School of Kálmán Laki, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., H-4032 Debrecen, Hungary
| | - Tibor István Szük
- Department of Cardiology and Cardiac Surgery, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., H-4032 Debrecen, Hungary; Doctoral School of Kálmán Laki, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98., H-4032 Debrecen, Hungary.
| |
Collapse
|
39
|
Vorn R, Mithani S, Devoto C, Meier TB, Lai C, Yun S, Broglio SP, McAllister TW, Giza CC, Kim HS, Huber D, Harezlak J, Cameron KL, McGinty G, Jackson J, Guskiewicz KM, Mihalik JP, Brooks A, Duma S, Rowson S, Nelson LD, Pasquina P, McCrea MA, Gill JM. Proteomic Profiling of Plasma Biomarkers Associated With Return to Sport Following Concussion: Findings From the NCAA and Department of Defense CARE Consortium. Front Neurol 2022; 13:901238. [PMID: 35928129 PMCID: PMC9343581 DOI: 10.3389/fneur.2022.901238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/08/2022] [Indexed: 11/30/2022] Open
Abstract
Objective To investigate the plasma proteomic profiling in identifying biomarkers related to return to sport (RTS) following a sport-related concussion (SRC). Methods This multicenter, prospective, case-control study was part of a larger cohort study conducted by the NCAA-DoD Concussion Assessment, Research, and Education (CARE) Consortium, athletes (n = 140) with blood collected within 48 h of injury and reported day to asymptomatic were included in this study, divided into two groups: (1) recovery <14-days (n = 99) and (2) recovery ≥14-days (n = 41). We applied a highly multiplexed proteomic technique that uses DNA aptamers assay to target 1,305 proteins in plasma samples from concussed athletes with <14-days and ≥14-days. Results We identified 87 plasma proteins significantly dysregulated (32 upregulated and 55 downregulated) in concussed athletes with recovery ≥14-days relative to recovery <14-days groups. The significantly dysregulated proteins were uploaded to Ingenuity Pathway Analysis (IPA) software for analysis. Pathway analysis showed that significantly dysregulated proteins were associated with STAT3 pathway, regulation of the epithelial mesenchymal transition by growth factors pathway, and acute phase response signaling. Conclusion Our data showed the feasibility of large-scale plasma proteomic profiling in concussed athletes with a <14-days and ≥ 14-days recovery. These findings provide a possible understanding of the pathophysiological mechanism in neurobiological recovery. Further study is required to determine whether these proteins can aid clinicians in RTS decisions.
Collapse
Affiliation(s)
- Rany Vorn
- School of Nursing, Johns Hopkins University, Baltimore, MD, United States
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| | - Sara Mithani
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States
- School of Nursing, University of Texas Health Science Center San Antonio, San Antonio, TX, United States
| | - Christina Devoto
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Timothy B. Meier
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Chen Lai
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Sijung Yun
- Predictiv Care, Mountain View, CA, United States
| | - Steven P. Broglio
- Michigan Concussion Center, University of Michigan, Ann Arbor, MI, United States
| | - Thomas W. McAllister
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Christopher C. Giza
- Departments of Pediatrics and Neurosurgery, University of California, Los Angeles, Los Angeles, CA, United States
- UCLA Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Hyung-Suk Kim
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| | - Daniel Huber
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jaroslaw Harezlak
- Department of Epidemiology and Biostatistics School of Public Health-Bloomington, Indiana University, Bloomington, IN, United States
| | - Kenneth L. Cameron
- John A. Feagin Sports Medicine Fellowship, Keller Army Hospital, West Point, NY, United States
| | - Gerald McGinty
- United States Air Force Academy, Colorado Springs, CO, United States
| | - Jonathan Jackson
- United States Air Force Academy, Colorado Springs, CO, United States
| | - Kevin M. Guskiewicz
- Matthew Gfeller Center, Department of Exercise and Sport Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Jason P. Mihalik
- Matthew Gfeller Center, Department of Exercise and Sport Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Alison Brooks
- Department of Orthopedics, Division of Sports Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Stefan Duma
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, United States
| | - Steven Rowson
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, United States
| | - Lindsay D. Nelson
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Paul Pasquina
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Michael A. McCrea
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jessica M. Gill
- School of Nursing, Johns Hopkins University, Baltimore, MD, United States
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Neurology, Johns Hopkins University, Baltimore, MD, United States
- *Correspondence: Jessica M. Gill
| |
Collapse
|
40
|
Effect of Low High-Density Lipoprotein Level on Endothelial Activation and Prothrombotic Processes in Coronary Artery Disease-A Pilot Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19148637. [PMID: 35886486 PMCID: PMC9316205 DOI: 10.3390/ijerph19148637] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 11/17/2022]
Abstract
High-density lipoproteins (HDL) play an important role in the prevention of atherosclerosis. The aim of the study was to assess the relationship between serum HDL-C concentration and proinflammatory/prothrombic activation in coronary artery disease (CAD) patients. The study group included 27 acute myocardial infarction (AMI) patients and 30 stable angina pectoris (SA) patients. The control group consisted of 23 people without cardiac symptoms. In the AMI and SA groups, a lower HDL-C and a higher LDL-C/HDL-C index were observed. The SA patients had lower total cholesterol, LDL-C, sE-selectin ligand, as well as higher triglycerides and CD40 concentration in comparison with both the control and AMI groups. A higher von Willebrand Factor and intercellular adhesion molecule-1 were found in both study groups. Low HDL-C concentration in the CAD patients may intensify pro-inflammatory endothelial activation and prothrombotic processes. A low concentration of HDL-C and a high value of the LDL-C/HDL-C index seem to be better indices of atherogenic processes than the LDL-C concentration alone.
Collapse
|
41
|
Extracellular Vesicles as an Index for Endothelial Injury and Cardiac Dysfunction in a Rodent Model of GDM. Int J Mol Sci 2022; 23:ijms23094970. [PMID: 35563365 PMCID: PMC9101204 DOI: 10.3390/ijms23094970] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/20/2022] [Accepted: 04/26/2022] [Indexed: 02/08/2023] Open
Abstract
Gestational diabetes mellitus (GDM) increases risk of adverse pregnancy outcomes and maternal cardiovascular complications. It is widely believed that maternal endothelial dysfunction is a critical determinant of these risks, however, connections to maternal cardiac dysfunction and mechanisms of pathogenesis are unclear. Circulating extracellular vesicles (EVs) are emerging biomarkers that may provide insights into the pathogenesis of GDM. We examined the impact of GDM on maternal cardiac and vascular health in a rat model of diet-induced obesity-associated GDM. We observed a >3-fold increase in circulating levels of endothelial EVs (p < 0.01) and von Willebrand factor (p < 0.001) in GDM rats. A significant increase in mitochondrial DNA (mtDNA) within circulating extracellular vesicles was also observed suggesting possible mitochondrial dysfunction in the vasculature. This was supported by nicotinamide adenine dinucleotide deficiency in aortas of GDM mice. GDM was also associated with cardiac remodeling (increased LV mass) and a marked impairment in maternal diastolic function (increased isovolumetric relaxation time [IVRT], p < 0.01). Finally, we observed a strong positive correlation between endothelial EV levels and IVRT (r = 0.57, p < 0.05). In summary, we observed maternal vascular and cardiac dysfunction in rodent GDM accompanied by increased circulating endothelial EVs and EV-associated mitochondrial DNA. Our study highlights a novel method for assessment of vascular injury in GDM and highlights vascular mitochondrial injury as a possible therapeutic target.
Collapse
|
42
|
The Role of von Willebrand Factor Antigen in Predicting Survival of Patients with HBV-Related Cirrhosis. Can J Gastroenterol Hepatol 2022; 2022:9035971. [PMID: 35360443 PMCID: PMC8964228 DOI: 10.1155/2022/9035971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 02/20/2022] [Accepted: 03/01/2022] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE The model for end-stage liver disease (MELD) scoring system cannot be used to assess the deterioration of patients with liver cirrhosis caused by infection and portal hypertension. Elevated von Willebrand factor antigen (vWF-Ag) in plasma is associated with portal pressure and complications in patients with liver cirrhosis. We aimed to evaluate whether the addition of vWF-Ag can improve the risk prediction ability of the MELD scoring system. METHODS A total of 228 patients with hepatitis B virus (HBV)-related liver cirrhosis were eligible for inclusion in this retrospective study. The vWF-Ag level was assessed by enzyme-linked immunosorbent assay (ELISA). The endpoint of this study was defined as the time to liver transplantation or death. Univariate and multivariate analyses were performed to assess the risk factors associated with transplant-free mortality. Receiver operating characteristic (ROC) curve analysis was used to assess potential discriminatory variables for transplant-free mortality. RESULTS During a median follow-up interval of 30.23 months, 124 patients (54.4%) reached the endpoint of this study. Patients who died or underwent liver transplantation had elevated levels of MELD and vWF-Ag. Moreover, vWF-Ag and MELD showed comparable predictive potential for transplant-free survival (area under the curve [AUC], vWF-Ag = 0.71; AUC, MELD = 0.73). Ultimately, vWF-Ag can significantly improve the predictive potential of MELD in determining transplant-free mortality (AUC, MELD-vWF-Ag = 0.79, P = 0.006). CONCLUSION An elevated vWF-Ag level was independently associated with transplant-free mortality in patients with liver cirrhosis. The inclusion of vWF-Ag in the MELD scoring system can improve mortality predictions in patients with liver cirrhosis.
Collapse
|
43
|
Michels A, Lillicrap D, Yacob M. Role of von Willebrand factor in venous thromboembolic disease. JVS Vasc Sci 2022; 3:17-29. [PMID: 35028601 PMCID: PMC8739873 DOI: 10.1016/j.jvssci.2021.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 08/23/2021] [Indexed: 02/07/2023] Open
Abstract
Objective Evolving evidence of the shared risk factors and pathogenic mechanisms in arterial and venous thrombosis questions of the strict vascular dichotomy of arterial vs venous. The connection between arterial and venous thrombosis has been highlighted by common underlying inflammatory processes, a concept known as thromboinflammatory disease. Using this relationship, we can apply knowledge from arterial disease to better understand and potentially mitigate venous disease. A protein that has been extensively studied in atherothrombotic disease and inflammation is von Willebrand factor (VWF). Because many predisposing and provoking factors of venous thromboembolism (VTE) have been shown to directly modulate VWF levels, it is, perhaps, not surprising that VWF has been highlighted by several recent association studies of patients with VTE. Methods In the present narrative review, we investigated more deeply the effects of VWF in venous disease by synthesizing the data from clinical studies of deep vein thrombosis of the limbs, pulmonary embolism, portal and cerebral vein thrombosis, and the complications of thrombosis, including post-thrombotic syndrome, venous insufficiency, and chronic thromboembolic pulmonary hypertension. We have also discussed the findings from preclinical studies to highlight novel VWF biochemistry in thrombosis and therapeutics. Results Across the spectrum of venous thromboembolic disease, we consistently observed that elevated VWF levels conferred an increased risk of VTE and long-term venous complications. We have highlighted important findings from VWF molecular research and have proposed mechanisms by which VWF participates in venous disease. Emerging evidence from preclinical studies might reveal novel targets for thromboinflammatory disease, including specific VWF pathophysiology. Furthermore, we have highlighted the utility of measuring VWF to prognosticate and risk stratify for VTE and its complications. Conclusions As the prevalence of inflammatory processes, such as aging, obesity, and diabetes increases in our population, it is critical to understand the evolving role of VWF in venous disease to guide clinical decisions and therapeutics.
Collapse
Affiliation(s)
- Alison Michels
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada.,Division of Cardiovascular Surgery, Queen's University, Kingston Health Sciences Centre, Kingston, Ontario, Canada
| | - David Lillicrap
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Michael Yacob
- Division of Cardiovascular Surgery, Queen's University, Kingston Health Sciences Centre, Kingston, Ontario, Canada
| |
Collapse
|
44
|
Xu L, Qiu Y, Li Y, Wei Y, Wan Y, Deng W. Tissue dynamics of von Willebrand factor characterized by a novel fluorescent protein-von Willebrand factor chimera. J Thromb Haemost 2022; 20:208-221. [PMID: 34592034 DOI: 10.1111/jth.15542] [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: 05/13/2021] [Revised: 09/23/2021] [Accepted: 09/27/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Tissue dynamics of von Willebrand factor (VWF) that are vital to its biological function have not been fully characterized. OBJECTIVE To develop a new fluorescent protein--VWF chimera (FP-VWF) that has similar hematologic function to wild-type VWF and use it to monitor the tissue dynamics of VWF distribution. METHODS Genotyping, platelet counting, tail bleeding time assay, agarose gels, western blot, platelet aggregation, proteolytic analysis, and ELISA were applied in characterizing the function of FP-VWF; fluorescence spectrometer and confocal fluorescence microscope were used to monitor the plasma and tissue distribution of FP-VWF. RESULTS The transgenic mice that carry the FP-VWF retain hematologic activity of VWF with plasma levels of FP-VWF reduced by 50% and there are reduced high molecular weight FP-VWF multimers compared to the wild-type mice. The GPIb-binding and ADAMTS-13 (A Disintegrin and Metalloprotease with ThrombSpondin type 1 motif, member 13) proteolytic efficiency of FP-VWF are similar to wild-type VWF. The tissue distribution of FP-VWF was probed directly through its intrinsic fluorescence at normal or stimulated status, which indicated that the medicine-stimulated endogenous FP-VWF seems primarily released from the aorta and cleared in the spleen. Similar results were observed in non-fluorescent mice through a standard immunofluorescence approach. The fluorescence signals of FP-VWF were also similar to the standard dye-based approach in detecting the FeCl3 -induced blood clotting in vivo. CONCLUSIONS Together, these results suggest that this novel FP-VWF chimera is valuable in probing the tissue dynamics of VWF in quite a few biological and pharmaceutical applications.
Collapse
Affiliation(s)
- Linru Xu
- Cyrus Tang Medical Institute and Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Yanyang Qiu
- Cyrus Tang Medical Institute and Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Yanqing Li
- Cyrus Tang Medical Institute and Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Yaxuan Wei
- Cyrus Tang Medical Institute and Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Yan Wan
- Cyrus Tang Medical Institute and Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Wei Deng
- Cyrus Tang Medical Institute and Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| |
Collapse
|
45
|
Udaya R, Sivakanesan R. Synopsis of Biomarkers of Atheromatous Plaque Formation, Rupture and Thrombosis in the Diagnosis of Acute Coronary Syndromes. Curr Cardiol Rev 2022; 18:53-62. [PMID: 35410616 PMCID: PMC9896418 DOI: 10.2174/1573403x18666220411113450] [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: 08/18/2021] [Revised: 12/08/2021] [Accepted: 01/15/2022] [Indexed: 11/22/2022] Open
Abstract
Acute coronary syndrome is the main cause of mortality and morbidity worldwide and early diagnosis is a challenge for clinicians. Though cardiac Troponin, the most commonly used biomarker, is the gold standard for myocardial necrosis, it is blind for ischemia without necrosis. Therefore, ideal biomarkers are essential in the care of patients presenting with symptoms suggestive of cardiac ischemia. The ideal biomarker or group of biomarkers of atheromatous plaque formation, rupture and thrombosis for timely and accurate diagnosis of acute coronary syndrome is a current need. Therefore, we discuss the existing understanding and future of biomarkers of atheromatous plaque formation, rupture and thrombosis of acute coronary syndrome in this review. Keywords were searched from Medline, ISI, IBSS and Google Scholar databases. Further, the authors conducted a manual search of other relevant journals and reference lists of primary articles. The development of high-sensitivity troponin assays facilitates earlier exclusion of acute coronary syndrome, contributing to a reduced length of stay at the emergency department, and earlier treatment resulting in better outcomes. Although researchers have investigated biomarkers of atheromatous plaque formation, rupture and thrombosis to help early diagnosis of cardiac ischemia, most of them necessitate validation from further analysis. Among these biomarkers, pregnancy-associated plasma protein-A, intercellular adhesion molecule-1, and endothelial cell-specific molecule- 1(endocan) have shown promising results in the early diagnosis of acute coronary syndrome but need further evaluation. However, the use of a combination of biomarkers representing varying pathophysiological mechanisms of cardiac ischemia will support risk assessment, diagnosis and prognosis in these patients and this is the way forward.
Collapse
Affiliation(s)
- Ralapanawa Udaya
- Address correspondence to this author at the Department of Medicine, University of Peradeniya, Galaha Rd, 20400, Sri Lanka; Tel: 0718495682; E-mail:
| | | |
Collapse
|
46
|
MicroRNA-320a-3p Signatures as a Satisfactory Predictor of Acute Coronary Syndrome and Attenuates Inflammation by Targeting X-Linked Inhibitor of Apoptosis Protein. Artery Res 2021. [DOI: 10.1007/s44200-021-00002-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
AbstractAcute coronary syndrome (ACS) is a heart disease with a high mortality rate. Recently, more and more evidence illustrated that microRNAs (miRNA) participated in regulating the occurrence of heart disease. This study aimed to detect the level of serum miR-320a-3p in patients with ACS, predict its possibility as a candidate gene for diagnosis, and explore its potential mechanism in the regulation of ACS. 139 ACS patients and 126 controls were recruited in this study. The expression level of miR-320a-3p was determined by qRT-PCR. The predictive value in ACS was assessed by receiver operating characteristic (ROC) curve. Enzyme-linked immunosorbent assay (ELISA) was used to measure the protein expression levels of inflammatory factors. The downstream targets of miR-320a-3p were verified by luciferase reporter gene assay. In ACS patients and rat models, the expression level of serum miR-320a-3p was significantly increased. ROC curve revealed that abnormal expression of miR-320a-3p was of diagnostic value for ACS. In an in vivo rat model, down-regulation of miR-320a-3p inhibited the production of von Willebrand factor (vWF), Heart fatty acid-binding protein (H-FABP), interleukin-1β (IL-1β), IL-6, and tumor necrosis factor-α (TNF-α). In other words, down-regulation of miR-320a-3p reduced rat vascular endothelial injury and inflammation. X-linked inhibitor of apoptosis protein (XIAP) was determined to be a direct target of miR-320a-3p. miR-320a-3p is useful for the diagnosis of ACS. Animal experiments confirmed that up-regulated miR-320a-3p promoted vascular endothelial injury and inflammatory response by targeting XIAP, thus promoting the development of ACS. MiR-320a-3p may be a new breakthrough in the diagnosis and treatment of ACS.
Collapse
|
47
|
Saeedi Saravi SS, Bonetti NR, Pugin B, Constancias F, Pasterk L, Gobbato S, Akhmedov A, Liberale L, Lüscher TF, Camici GG, Beer JH. Lifelong dietary omega-3 fatty acid suppresses thrombotic potential through gut microbiota alteration in aged mice. iScience 2021; 24:102897. [PMID: 34401676 PMCID: PMC8355916 DOI: 10.1016/j.isci.2021.102897] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/17/2021] [Accepted: 07/20/2021] [Indexed: 01/04/2023] Open
Abstract
Aging is a major risk factor for cardiovascular diseases, including thrombotic events. The gut microbiota has been implicated in the development of thrombotic risk. Plant-derived omega-3 fatty acid ɑ-linolenic acid (ALA) confers beneficial anti-platelet and anti-inflammatory effects. Hence, antithrombotic activity elicited by ALA may be partly dependent on its interaction with gut microbiota during aging. Here, we demonstrate that lifelong dietary ALA decreases platelet hyperresponsiveness and thrombus formation in aged mice. These phenotypic changes can be partly attributed to alteration of microbial composition and reduction of its metabolite trimethylamine N-oxide and inflammatory mediators including TNF-α, as well as the upregulated production of short-chain fatty acid acetate. ALA-rich diet also dampens secretion of increased procoagulant factors, tissue factor and plasminogen activator inhibitor-1, in aged mice. Our results suggest long-term ALA supplementation as an attractive, accessible, and well-tolerated nutritional strategy against age-associated platelet hyperreactivity and thrombotic potential.
Collapse
Affiliation(s)
- Seyed Soheil Saeedi Saravi
- Laboratory for Platelet Research, Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland
- Department of Internal Medicine, Cantonal Hospital Baden, Im Ergel 1, 5404 Baden, Switzerland
| | - Nicole R. Bonetti
- Laboratory for Platelet Research, Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland
- Department of Internal Medicine, Cantonal Hospital Baden, Im Ergel 1, 5404 Baden, Switzerland
| | - Benoit Pugin
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| | - Florentin Constancias
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| | - Lisa Pasterk
- Laboratory for Platelet Research, Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland
| | - Sara Gobbato
- Department of Internal Medicine, Cantonal Hospital Baden, Im Ergel 1, 5404 Baden, Switzerland
| | - Alexander Akhmedov
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland
| | - Luca Liberale
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Thomas F. Lüscher
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland
- Royal Brompton and Harefield Hospitals and Imperial College, London, UK
| | - Giovanni G. Camici
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland
- University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
- Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Jürg H. Beer
- Laboratory for Platelet Research, Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland
- Department of Internal Medicine, Cantonal Hospital Baden, Im Ergel 1, 5404 Baden, Switzerland
| |
Collapse
|
48
|
Tan CMJ, Lewandowski AJ, Williamson W, Huckstep OJ, Yu GZ, Fischer R, Simon JN, Alsharqi M, Mohamed A, Leeson P, Bertagnolli M. Proteomic Signature of Dysfunctional Circulating Endothelial Colony-Forming Cells of Young Adults. J Am Heart Assoc 2021; 10:e021119. [PMID: 34275329 PMCID: PMC8475699 DOI: 10.1161/jaha.121.021119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022]
Abstract
Background A subpopulation of endothelial progenitor cells called endothelial colony-forming cells (ECFCs) may offer a platform for cellular assessment in clinical studies because of their remarkable angiogenic and expansion potentials in vitro. Despite endothelial cell function being influenced by cardiovascular risk factors, no studies have yet provided a comprehensive proteomic profile to distinguish functional (ie, more angiogenic and expansive cells) versus dysfunctional circulating ECFCs of young adults. The aim of this study was to provide a detailed proteomic comparison between functional and dysfunctional ECFCs. Methods and Results Peripheral blood ECFCs were isolated from 11 subjects (45% men, aged 27±5 years) using Ficoll density gradient centrifugation. ECFCs expressed endothelial and progenitor surface markers and displayed cobblestone-patterned morphology with clonal and angiogenic capacities in vitro. ECFCs were deemed dysfunctional if <1 closed tube formed during the in vitro tube formation assay and proliferation rate was <20%. Hierarchical functional clustering revealed distinct ECFC proteomic signatures between functional and dysfunctional ECFCs with changes in cellular mechanisms involved in exocytosis, vesicle transport, extracellular matrix organization, cell metabolism, and apoptosis. Targeted antiangiogenic proteins in dysfunctional ECFCs included SPARC (secreted protein acidic and rich in cysteine), CD36 (cluster of differentiation 36), LUM (lumican), and PTX3 (pentraxin-related protein PYX3). Conclusions Circulating ECFCs with impaired angiogenesis and expansion capacities have a distinct proteomic profile and significant phenotype changes compared with highly angiogenic endothelial cells. Impaired angiogenesis in dysfunctional ECFCs may underlie the link between endothelial dysfunction and cardiovascular disease risks in young adults.
Collapse
Affiliation(s)
- Cheryl M. J. Tan
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
| | - Adam J. Lewandowski
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
| | - Wilby Williamson
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
| | - Odaro J. Huckstep
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
- Department of BiologyUnited States Air Force AcademyColorado SpringsCOUSA
| | - Grace Z. Yu
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
- Wellcome Centre for Human GeneticsUniversity of OxfordOxfordUK
| | - Roman Fischer
- Target Discovery Institute (TDI) Mass Spectrometry Laboratory, Target Discovery Institute, Nuffield Department of MedicineUniversity of OxfordOxfordUK
| | - Jillian N. Simon
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUnited Kingdom
| | - Maryam Alsharqi
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
- Department of Cardiac TechnologyImam Abdulrahman Bin Faisal UniversityDammamSaudi Arabia
| | - Afifah Mohamed
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
- Department of Diagnostic Imaging & Applied Health Sciences, Faculty of Health SciencesUniversiti Kebangsaan MalaysiaKuala LumpurMalaysia
| | - Paul Leeson
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
| | - Mariane Bertagnolli
- Oxford Cardiovascular Clinical Research Facility, Radcliffe Department of Medicine, Division of Cardiovascular MedicineUniversity of OxfordOxfordUK
- Montreal Hospital Sacré‐Cœur Research CentreCentre Intégré Universitaire de Santé et de Services Sociaux du Nord‐de‐l'Île‐de‐MontréalMontréalQCCanada
- School of Physical and Occupational Therapy, Faculty of MedicineMcGill UniversityMontréalQCCanada
| |
Collapse
|
49
|
Todorov SS, Deribas VJ, Kazmin AS, Todorov SS. [Morphological and Molecular-Biological Changes in the Coronary Arteries after Stenting]. ACTA ACUST UNITED AC 2021; 61:79-84. [PMID: 34397345 DOI: 10.18087/cardio.2021.7.n1211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/10/2020] [Accepted: 07/29/2020] [Indexed: 11/18/2022]
Abstract
This review addresses morphological changes in coronary arteries following stenting, which result from damage to the vascular wall. These changes include 1) formation of a thrombus in the site of intimal injury; 2) inflammation; 3) proliferation and migration of smooth muscle cells; 4) formation of extracellular matrix. Each of these pathological processes has specific morpho-biological features. The review shows the role of von Willebrand factor in development of early thrombosis after intimal injury, which provokes activation of the inflammatory response followed by proliferation of smooth muscle cell that synthetize the extracellular matrix. These cellular and intercellular changes are based on overexpression of TGF-β1 protein, which facilitates modulation of various types of smooth muscle cells, including contractile and secretory ones. Issues of fine regulation of cellular and intercellular interactions by apoptosis, activation of mTOR signaling molecules, and microRNA are still understudied. Dynamic changes in drug-coated stents during development of neoatherosclerosis and late thrombosis remain not elucidated. Current reports show that initial mechanisms triggering pathological regenerative and hyperplastic processes that result in coronary restenosis in the area of implanted stents may form early (first hours or days) after stenting. Most studies were performed on experimental rather than on autopsy material, which does not allow fully unbiased interpretation of obtained data. Studying dynamics of morphological and molecular changes in coronary arteries after stenting, including on autopsy material, will allow one to express an opinion on the risk of postoperative thrombosis and restenosis.
Collapse
Affiliation(s)
- S S Todorov
- Rostov State Medical University of the Ministry of Health of Russia, Rostov-on-Don
| | - V J Deribas
- Rostov State Medical University of the Ministry of Health of Russia, Rostov-on-Don
| | - A S Kazmin
- Rostov State Medical University of the Ministry of Health of Russia, Rostov-on-Don
| | - S S Todorov
- Rostov State Medical University of the Ministry of Health of Russia, Rostov-on-Don
| |
Collapse
|
50
|
Nitter TB, Hilt B, Svendsen KVH, Buhagen M, Jørgensen RB. Association between exposure to different stone aggregates from asphalt and blood coagulability: A human exposure chamber study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 778:146309. [PMID: 33714824 DOI: 10.1016/j.scitotenv.2021.146309] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/26/2021] [Accepted: 03/02/2021] [Indexed: 06/12/2023]
Abstract
A large fraction of particulate matter (PM), especially PM10, concentrations are due to non-exhaust emissions, such as road abrasion and wear on tires and brake pads. Concentrating on road abrasion, we aimed to investigate blood coagulability in healthy adults after exposure to two types of stone materials commonly used in asphalt on Norwegian roads. This study followed a randomized, double-blind, cross-over study design. Using an exposure chamber, 24 healthy young volunteers were exposed to aggregates of two different types of rocks and placebo dust: quartz diorite, rhomb porphyry, and lactose (placebo dust). Each exposure session lasted for 4 hours (h), and blood samples were collected before exposure (baseline), 4 h post-exposure, and 24 h post-exposure to analyse potential changes in the von Willebrand factor (vWF) as well as of fibrinogen, d-dimer, leukocytes, and thrombocytes. The dust concentration in the exposure chamber was measured with real-time instruments and gravimetric samples of total dust, respirable dust, PM10, PM2.5, and ultrafine particles (UFP). The results were analysed using a linear mixed-effect model. Leukocyte blood counts increased post-exposure for all exposure materials; however, none of the increases were statistically significant. The concentration of fibrinogen increased after exposure to quartz diorite, while it decreased after exposures to rhomb porphyry and lactose. Type of material was a statistically significant explanatory variable for the concentration of fibrinogen, with the most significant increase occurring 24 h post-exposure to quartz diorite. After exposure to the three materials, vWF decreased. For the thrombocytes, an increase in blood count was observed 24 h post-exposure to quartz diorite and rhomb porphyry, with a modest (p = 0.09) positive association for quartz diorite. Although the results are limited, we conclude that the different effects observed post-exposure to quartz diorite support considering potential health effects when choosing materials in the production of asphalt.
Collapse
Affiliation(s)
- Therese Bergh Nitter
- Department of Industrial Economics and Technology Management, Norwegian University of Science and Technology (NTNU), Norway.
| | - Bjørn Hilt
- Department of Occupational Medicine, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway; Department of Public Health and Nursing, NTNU, Norway
| | - Kristin V Hirsch Svendsen
- Department of Industrial Economics and Technology Management, Norwegian University of Science and Technology (NTNU), Norway
| | - Morten Buhagen
- Department of Occupational Medicine, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway; Department of Public Health and Nursing, NTNU, Norway
| | - Rikke Bramming Jørgensen
- Department of Industrial Economics and Technology Management, Norwegian University of Science and Technology (NTNU), Norway
| |
Collapse
|