1
|
D'Italia G, Schroen B, Cosemans JM. Commonalities of platelet dysfunction in heart failure with preserved ejection fraction and underlying comorbidities. ESC Heart Fail 2025; 12:1013-1028. [PMID: 39375979 PMCID: PMC11911585 DOI: 10.1002/ehf2.15090] [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: 06/10/2024] [Revised: 08/06/2024] [Accepted: 09/06/2024] [Indexed: 10/09/2024] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is characterized by a lack of a specific targeted treatment and a complex, partially unexplored pathophysiology. Common comorbidities associated with HFpEF are hypertension, atrial fibrillation, obesity and diabetes. These comorbidities, combined with advanced age, play a crucial role in the initiation and development of the disease through the promotion of systemic inflammation and consequent changes in cardiac phenotype. In this context, we suggest platelets as important players due to their emerging role in vascular inflammation. This review provides an overview of the role of platelets in HFpEF and its associated comorbidities, including hypertension, atrial fibrillation, obesity and diabetes mellitus, as well as the impact of age and sex on platelet function. These major HFpEF-associated comorbidities present alterations in platelet behaviour and in features linked to platelet size, content and reactivity. The resulting dysfunctional platelets can contribute to further increase inflammation, oxidative stress and endothelial dysfunction, suggesting an active role of these cells in the initiation and progression of HFpEF. Recent evidence shows that reduced platelet count and elevated mean platelet volume are associated with worsening heart failure in HFpEF patients. However, the specific mechanisms by which platelets contribute to HFpEF development and progression are still largely unexplored, with only a few studies investigating platelet function in HFpEF. We discuss the limited yet significant body of research investigating platelet function in HFpEF, emphasizing the need for more comprehensive studies. Additionally, we explore the potential mechanisms through which platelets may influence HFpEF, such as their interactions with the vascular endothelium and the secretion of bioactive molecules like cytokines, chemokines and RNA molecules. These interactions and secretions may play a role in modulating vascular inflammation and contributing to the pathophysiological landscape of HFpEF. The review underscores the necessity for future research to elucidate the precise contributions of platelets to HFpEF, aiming to potentially identify novel therapeutic targets and improve patient outcomes. The evidence presented herein supports the hypothesis that platelets are not merely passive bystanders but active participants in the pathophysiology of HFpEF and its comorbidities.
Collapse
Affiliation(s)
- Giorgia D'Italia
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
| | - Blanche Schroen
- Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
| | - Judith M.E.M. Cosemans
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtThe Netherlands
| |
Collapse
|
2
|
Mortezaei A, Ghorbani M, Hajikarimloo B, Sameer O, Kazemi T, Salavati E, Hamidpour M, Gheydari ME. Is L-Arginine an Appropriate Alternative for Conventional Anti-Atherosclerotic Therapy?: A Comprehensive Review. Health Sci Rep 2025; 8:e70544. [PMID: 40161001 PMCID: PMC11949766 DOI: 10.1002/hsr2.70544] [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: 09/19/2024] [Revised: 01/31/2025] [Accepted: 02/14/2025] [Indexed: 04/02/2025] Open
Abstract
Background Atherosclerosis is the leading cause of cardiovascular disease (CVD). Historically, the management of atherosclerosis was focused on decreasing lipid profile levels; however, recent evidence demonstrated that platelets and leukocytes play an important role in forming and exacerbating atherosclerosis. L-arginine (L-Arg), a precursor to nitric oxide (NO), plays a critical role in modulating oxidative stress and influencing platelet-leukocyte recruitment and has been extensively addressed in the context of CVD. Objective We aimed to perform a comprehensive literature review on l-Arg metabolism in the causative pathway of atherosclerosis compared to conventional treatment and it as a putative therapeutic approach. Results L-Arg supplementation has shown promising effects on NO production, improving endothelial function and reducing oxidative stress in preclinical models. Clinical studies have indicated moderate improvements in vascular health markers, including reductions in inflammation and oxidative stress, although results have varied across studies. The potential of l-Arg to modify platelet-leukocyte recruitment and slow the progression of atherosclerotic plaque development has been observed in certain studies. However, these benefits remain inconsistent, and more robust clinical trials are needed to confirm its effectiveness. Additionally, while l-Arg appears to be relatively safe, some studies reported mild gastrointestinal discomfort as a common side effect. Conclusion l-Arg holds potential as a complementary or alternative treatment for atherosclerosis, particularly in improving endothelial function and reducing inflammation and oxidative stress. However, the variability in clinical outcomes and the lack of long-term data required further investigation into assessing therapeutic benefits. Future studies should focus on determining optimal dosing regimens, evaluating their long-term safety, and assessing their potential in combination with other therapies to enhance cardiovascular outcomes.
Collapse
Affiliation(s)
- Ali Mortezaei
- Student Research CommitteeGonabad University of Medical SciencesGonabadIran
| | - Mohammad Ghorbani
- Faculty of Allied Medicine, Department of Medical Laboratory SciencesGonabad University of Medical SciencesGonabadIran
- Department of Hematology and Blood Banking, School of Allied Medical SciencesShahid Beheshti University of Medical SciencesTehranIran
| | | | - Omar Sameer
- College of MedicineUniversity of SharjahSharjahUAE
| | - Toba Kazemi
- Cardiovascular Diseases Research CenterBirjand University of Medical SciencesBirjandIran
| | - Ebrahim Salavati
- Allameh Bohlool HospitalGonabad University of Medical SciencesGonabadIran
| | - Mohsen Hamidpour
- HSC Research Center, Department of Hematology and Blood Banking, School of Allied Medical SciencesShahid Beheshti University of Medical SciencesTehranIran
| | - Mohammad Esmail Gheydari
- Department of Cardiology, School of Medicine, Taleghani HospitalShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
3
|
Paolini L, Guida F, Calvaruso A, Andreozzi L, Pierantoni L, Lanari M, Fabi M. Endothelial Dysfunction: Molecular Mechanisms and Therapeutic Strategies in Kawasaki Disease. Int J Mol Sci 2024; 25:13322. [PMID: 39769085 PMCID: PMC11676170 DOI: 10.3390/ijms252413322] [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: 10/30/2024] [Revised: 11/30/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
The endothelium plays a key role in regulating vascular homeostasis by responding to a large spectrum of chemical and physical stimuli. Vasculitis is a group of inflammatory conditions affecting the vascular bed, and it is known that they are strongly linked to endothelial dysfunction (ED). Kawasaki disease (KD) is one childhood systemic vasculitis, and it represents the leading cause of acquired cardiac disease in children due to coronary damage and subsequent cardiovascular (CV) morbidity and mortality. We aimed to focus on the actual knowledge of ED in the pathogenesis of KD and its practical implications on therapeutical strategies to limit cardiovascular complications. Understanding ED in KD provides insight into the underlying mechanisms and identifies potential therapeutic targets to mitigate vascular damage, ultimately improving cardiovascular outcomes in both the acute and chronic stages of the disease. However, research gaps remain, particularly in translating findings from animal models into clinical applications for cardiovascular lesions and related morbidity in KD patients.
Collapse
Affiliation(s)
- Lucia Paolini
- Specialty School of Paediatrics, Alma Mater Studiorum, University of Bologna, 40139 Bologna, Italy; (L.P.); (A.C.)
| | - Fiorentina Guida
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40139 Bologna, Italy; (F.G.); (L.A.); (L.P.); (M.L.)
| | - Antonino Calvaruso
- Specialty School of Paediatrics, Alma Mater Studiorum, University of Bologna, 40139 Bologna, Italy; (L.P.); (A.C.)
| | - Laura Andreozzi
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40139 Bologna, Italy; (F.G.); (L.A.); (L.P.); (M.L.)
| | - Luca Pierantoni
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40139 Bologna, Italy; (F.G.); (L.A.); (L.P.); (M.L.)
| | - Marcello Lanari
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40139 Bologna, Italy; (F.G.); (L.A.); (L.P.); (M.L.)
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, 40139 Bologna, Italy
| | - Marianna Fabi
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40139 Bologna, Italy; (F.G.); (L.A.); (L.P.); (M.L.)
| |
Collapse
|
4
|
Nguyen BA, Alexander MR, Harrison DG. Immune mechanisms in the pathophysiology of hypertension. Nat Rev Nephrol 2024; 20:530-540. [PMID: 38658669 DOI: 10.1038/s41581-024-00838-w] [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] [Accepted: 04/03/2024] [Indexed: 04/26/2024]
Abstract
Hypertension is a leading risk factor for morbidity and mortality worldwide. Despite current anti-hypertensive therapies, most individuals with hypertension fail to achieve adequate blood pressure control. Moreover, even with adequate control, a residual risk of cardiovascular events and associated organ damage remains. These findings suggest that current treatment modalities are not addressing a key element of the underlying pathology. Emerging evidence implicates immune cells as key mediators in the development and progression of hypertension. In this Review, we discuss our current understanding of the diverse roles of innate and adaptive immune cells in hypertension, highlighting key findings from human and rodent studies. We explore mechanisms by which these immune cells promote hypertensive pathophysiology, shedding light on their multifaceted involvement. In addition, we highlight advances in our understanding of autoimmunity, HIV and immune checkpoints that provide valuable insight into mechanisms of chronic and dysregulated inflammation in hypertension.
Collapse
Affiliation(s)
- Bianca A Nguyen
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Matthew R Alexander
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, TN, USA
| | - David G Harrison
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Institute for Infection, Immunology and Inflammation, Nashville, TN, USA.
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
5
|
Chandy M, Hill T, Jimenez-Tellez N, Wu JC, Sarles SE, Hensel E, Wang Q, Rahman I, Conklin DJ. Addressing Cardiovascular Toxicity Risk of Electronic Nicotine Delivery Systems in the Twenty-First Century: "What Are the Tools Needed for the Job?" and "Do We Have Them?". Cardiovasc Toxicol 2024; 24:435-471. [PMID: 38555547 PMCID: PMC11485265 DOI: 10.1007/s12012-024-09850-9] [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: 12/07/2023] [Accepted: 03/19/2024] [Indexed: 04/02/2024]
Abstract
Cigarette smoking is positively and robustly associated with cardiovascular disease (CVD), including hypertension, atherosclerosis, cardiac arrhythmias, stroke, thromboembolism, myocardial infarctions, and heart failure. However, after more than a decade of ENDS presence in the U.S. marketplace, uncertainty persists regarding the long-term health consequences of ENDS use for CVD. New approach methods (NAMs) in the field of toxicology are being developed to enhance rapid prediction of human health hazards. Recent technical advances can now consider impact of biological factors such as sex and race/ethnicity, permitting application of NAMs findings to health equity and environmental justice issues. This has been the case for hazard assessments of drugs and environmental chemicals in areas such as cardiovascular, respiratory, and developmental toxicity. Despite these advances, a shortage of widely accepted methodologies to predict the impact of ENDS use on human health slows the application of regulatory oversight and the protection of public health. Minimizing the time between the emergence of risk (e.g., ENDS use) and the administration of well-founded regulatory policy requires thoughtful consideration of the currently available sources of data, their applicability to the prediction of health outcomes, and whether these available data streams are enough to support an actionable decision. This challenge forms the basis of this white paper on how best to reveal potential toxicities of ENDS use in the human cardiovascular system-a primary target of conventional tobacco smoking. We identify current approaches used to evaluate the impacts of tobacco on cardiovascular health, in particular emerging techniques that replace, reduce, and refine slower and more costly animal models with NAMs platforms that can be applied to tobacco regulatory science. The limitations of these emerging platforms are addressed, and systems biology approaches to close the knowledge gap between traditional models and NAMs are proposed. It is hoped that these suggestions and their adoption within the greater scientific community will result in fresh data streams that will support and enhance the scientific evaluation and subsequent decision-making of tobacco regulatory agencies worldwide.
Collapse
Affiliation(s)
- Mark Chandy
- Robarts Research Institute, Western University, London, N6A 5K8, Canada
| | - Thomas Hill
- Division of Nonclinical Science, Center for Tobacco Products, US Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Nerea Jimenez-Tellez
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94304, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94304, USA
| | - S Emma Sarles
- Biomedical and Chemical Engineering PhD Program, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Edward Hensel
- Department of Mechanical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Qixin Wang
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Daniel J Conklin
- Division of Environmental Medicine, Department of Medicine, Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville, 580 S. Preston St., Delia Baxter, Rm. 404E, Louisville, KY, 40202, USA.
| |
Collapse
|
6
|
Tomidy J, Satriadinatha GBY, Liwang FK, Maharani K, Imran D, Estiasari R. Prognostic identifier of cerebrovascular complications in tuberculous meningitis: Meta-analysis. J Stroke Cerebrovasc Dis 2023; 32:107371. [PMID: 37738916 DOI: 10.1016/j.jstrokecerebrovasdis.2023.107371] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 09/03/2023] [Accepted: 09/12/2023] [Indexed: 09/24/2023] Open
Abstract
INTRODUCTION Cerebrovascular complications could occur in 15-57 % of patients with tuberculous meningitis (TBM). It is crucial to rapidly identify TBM patients who are at risk for stroke. This study aimed to find predictors of stroke in patients with TBM. METHODS This systematic review and meta-analysis were done using literature searches through online databases up to April 30th, 2022. Three independent authors performed literature screening, data extraction, and critical appraisal of the studies. Eight studies involving 1535 samples were included. RESULTS We analyzed data regarding demographic, comorbidity, clinical presentation, radiologic, and laboratory parameters. Overall, clinical presentation that showed outcome difference was found in patients with findings of vomiting (OR = 2.71, 95 % CI: 1.30-5.63), cranial nerve deficit (OR = 4.10, 95 % CI: 1.83-9.21), focal deficit (OR = 5.56, 95 % CI: 2.24-13.79), and altered consciousness (OR = 1.90, 95 % CI: 1.24-2.92). Some comorbidities showed significant differences such as diabetes mellitus (OR = 2.58, 95 % CI: 1.51-4.41), hypertension (OR = 5.73, 95 % CI: 3.36-9.77), ischemic heart disease (OR = 2.18, 95 % CI: 1.02-4.63), and smoking (OR = 2.65, 95 % CI: 1.22-5.77). Two radiological changes shown to have significantly higher proportions are hydrocephalus (OR = 2.50, 95 % CI: 1.74-3.58) and meningeal enhancements (OR = 3.99, 95 % CI: 1.73-9.20). CONCLUSION Our analysis indicated that clinical presentations of vomiting, cranial nerve deficit, focal deficit, altered consciousness; comorbidity of diabetes mellitus, hypertension, smoking history, ischemic heart disease; and radiological findings of meningeal enhancement and hydrocephalus showed significant association with stroke incidence in tuberculous meningitis.
Collapse
Affiliation(s)
- Julianto Tomidy
- Department of Neurology, Cipto Mangunkusumo Hospital, Faculty of Medicine Universitas Indonesia, Jl. Diponegoro No.71, Jakarta 10430, Indonesia
| | - Gede Bagus Yoga Satriadinatha
- Department of Neurology, Cipto Mangunkusumo Hospital, Faculty of Medicine Universitas Indonesia, Jl. Diponegoro No.71, Jakarta 10430, Indonesia
| | - Filbert Kurnia Liwang
- Department of Neurology, Cipto Mangunkusumo Hospital, Faculty of Medicine Universitas Indonesia, Jl. Diponegoro No.71, Jakarta 10430, Indonesia
| | - Kartika Maharani
- Department of Neurology, Cipto Mangunkusumo Hospital, Faculty of Medicine Universitas Indonesia, Jl. Diponegoro No.71, Jakarta 10430, Indonesia
| | - Darma Imran
- Department of Neurology, Cipto Mangunkusumo Hospital, Faculty of Medicine Universitas Indonesia, Jl. Diponegoro No.71, Jakarta 10430, Indonesia
| | - Riwanti Estiasari
- Department of Neurology, Cipto Mangunkusumo Hospital, Faculty of Medicine Universitas Indonesia, Jl. Diponegoro No.71, Jakarta 10430, Indonesia.
| |
Collapse
|
7
|
Xu Y, Guo Y. Platelet indices and blood pressure: a multivariable mendelian randomization study. Thromb J 2023; 21:31. [PMID: 36941692 PMCID: PMC10026509 DOI: 10.1186/s12959-023-00475-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/10/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Platelet indices are blood-based parameters reflecting the activation of platelets. Previous studies have identified an association between platelet indices and blood pressure (BP). However, causal inferences are prone to bias by confounding effects and reverse causation. We performed a Mendelian randomization (MR) study to compare the causal roles between genetically determined platelet indices and BP levels. METHODS Single-nucleotide polymorphisms (SNPs) associated with platelet count (PLT), plateletcrit (PCT), mean platelet volume (MPV), platelet distribution width (PDW), and BP at the level of genome-wide significance (p < 5 × 10- 8) in the UK Biobank were used as instrumental variables. In bidirectional univariable MR analyses, inverse variance-weighted (IVW), MR‒Egger, and weighted median methods were used to obtain estimates for individual causal power. In addition, heterogeneity and sensitivity analyses were performed to examine the pleiotropy of effect estimates. Finally, multivariable MR analyses were undertaken to disentangle the comparative effects of four platelet indices on BP. RESULTS In the univariable MR analyses, increased levels of PLT and PCT were associated with higher BP, and PDW was associated with higher DBP alone. In the reverse direction, SBP had a minor influence on PLT and PCT. In multivariable MR analysis, PDW and PLT revealed an independent effect, whereas the association for PCT and MPV was insignificant after colinear correction. CONCLUSION These findings suggest that platelets and BP may affect each other. PDW and PLT are independent platelet indices influencing BP. Increased platelet activation and aggregation may be involved in the pathogenesis of hypertension, which may provide insights into evaluating thromboembolic events in people with high BP. The necessity of initiating antiplatelet therapy among hypertension groups needs further investigation.
Collapse
Affiliation(s)
- Yuhan Xu
- School of Medicine, Southeast University, Nanjing, Jiangsu Province, 210009, China
- Department of Neurology, Affiliated ZhongDa Hospital of Southeast University, Nanjing, China
| | - Yijing Guo
- School of Medicine, Southeast University, Nanjing, Jiangsu Province, 210009, China.
- Department of Neurology, Affiliated ZhongDa Hospital of Southeast University, Nanjing, China.
| |
Collapse
|
8
|
Rolling CC, Sowa MA, Wang TT, Cornwell M, Myndzar K, Schwartz T, El Bannoudi H, Buyon J, Barrett TJ, Berger JS. P2Y12 Inhibition Suppresses Proinflammatory Platelet-Monocyte Interactions. Thromb Haemost 2023; 123:231-244. [PMID: 36630990 PMCID: PMC11007758 DOI: 10.1055/s-0042-1758655] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Monocyte-platelet aggregates (MPAs) represent the crossroads between thrombosis and inflammation, and targeting this axis may suppress thromboinflammation. While antiplatelet therapy (APT) reduces platelet-platelet aggregation and thrombosis, its effects on MPA and platelet effector properties on monocytes are uncertain. OBJECTIVES To analyze the effect of platelets on monocyte activation and APT on MPA and platelet-induced monocyte activation. METHODS Agonist-stimulated whole blood was incubated in the presence of P-selectin, PSGL1, PAR1, P2Y12, GP IIb/IIIa, and COX-1 inhibitors and assessed for platelet and monocyte activity via flow cytometry. RNA-Seq of monocytes incubated with platelets was used to identify platelet-induced monocyte transcripts and was validated by RT-qPCR in monocyte-PR co-incubation ± APT. RESULTS Consistent with a proinflammatory platelet effector role, MPAs were increased in patients with COVID-19. RNA-Seq revealed a thromboinflammatory monocyte transcriptome upon incubation with platelets. Monocytes aggregated to platelets expressed higher CD40 and tissue factor than monocytes without platelets (p < 0.05 for each). Inhibition with P-selectin (85% reduction) and PSGL1 (87% reduction) led to a robust decrease in MPA. P2Y12 and PAR1 inhibition lowered MPA formation (30 and 21% reduction, p < 0.05, respectively) and decreased monocyte CD40 and TF expression, while GP IIb/IIIa and COX1 inhibition had no effect. Pretreatment of platelets with P2Y12 inhibitors reduced the expression of platelet-mediated monocyte transcription of proinflammatory SOCS3 and OSM. CONCLUSIONS: Platelets skew monocytes toward a proinflammatory phenotype. Among traditional APTs, P2Y12 inhibition attenuates platelet-induced monocyte activation.
Collapse
Affiliation(s)
- Christina C. Rolling
- Department of Medicine, New York University Grossman School of Medicine, New York, NY
- University Medical Center Hamburg-Eppendorf, Department of Oncology and Hematology, Hamburg, Germany
| | - Marcin A. Sowa
- Department of Medicine, New York University Grossman School of Medicine, New York, NY
| | - Tricia T. Wang
- Department of Medicine, New York University Grossman School of Medicine, New York, NY
| | - MacIntosh Cornwell
- Department of Medicine, New York University Grossman School of Medicine, New York, NY
| | - Khrystyna Myndzar
- Department of Medicine, New York University Grossman School of Medicine, New York, NY
| | - Tamar Schwartz
- Department of Medicine, New York University Grossman School of Medicine, New York, NY
| | - Hanane El Bannoudi
- Department of Medicine, New York University Grossman School of Medicine, New York, NY
| | - Jill Buyon
- Department of Medicine, New York University Grossman School of Medicine, New York, NY
| | - Tessa J. Barrett
- Department of Medicine, New York University Grossman School of Medicine, New York, NY
| | - Jeffrey S. Berger
- Department of Medicine, New York University Grossman School of Medicine, New York, NY
| |
Collapse
|
9
|
Farrah TE, Melville V, Czopek A, Fok H, Bruce L, Mills NL, Bailey MA, Webb DJ, Dear JW, Dhaun N. Arterial stiffness, endothelial dysfunction and impaired fibrinolysis are pathogenic mechanisms contributing to cardiovascular risk in ANCA-associated vasculitis. Kidney Int 2022; 102:1115-1126. [PMID: 35998848 DOI: 10.1016/j.kint.2022.07.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 06/20/2022] [Accepted: 07/11/2022] [Indexed: 12/14/2022]
Abstract
Cardiovascular disease is a complication of systemic inflammatory diseases including anti-neutrophil cytoplasm antibody-associated vasculitis (AAV). The mechanisms of cardiovascular morbidity in AAV are poorly understood, and risk-reduction strategies are lacking. Therefore, in a series of double-blind, randomized case-control forearm plethysmography and crossover systemic interventional studies, we examined arterial stiffness and endothelial function in patients with AAV in long-term disease remission and in matched healthy volunteers (32 each group). The primary outcome for the case-control study was the difference in endothelium-dependent vasodilation between health and AAV, and for the crossover study was the difference in pulse wave velocity (PWV) between treatment with placebo and selective endothelin-A receptor antagonism. Parallel in vitro studies of circulating monocytes and platelets explored mechanisms. Compared to healthy volunteers, patients with AAV had 30% reduced endothelium-dependent vasodilation and 50% reduced acute release of endothelial active tissue plasminogen activator (tPA), both significant in the case-control study. Patients with AAV had significantly increased arterial stiffness (PWV: 7.3 versus 6.4 m/s). Plasma endothelin-1 was two-fold higher in AAV and independently predicted PWV and tPA release. Compared to placebo, both selective endothelin-A and dual endothelin-A/B receptor blockade reduced PWV and increased tPA release in AAV in the crossover study. Mechanistically, patients with AAV had increased platelet activation, more platelet-monocyte aggregates, and altered monocyte endothelin receptor function, reflecting reduced endothelin-1 clearance. Patients with AAV in long-term remission have elevated cardiovascular risk and endothelin-1 contributes to this. Thus, our data support a role for endothelin-blockers to reduce cardiovascular risk by reducing arterial stiffness and increasing circulating tPA activity.
Collapse
Affiliation(s)
- Tariq E Farrah
- British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK; Clinical Research Centre, University of Edinburgh, Western General Hospital, Edinburgh, UK; Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Vanessa Melville
- Clinical Research Centre, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Alicja Czopek
- British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Henry Fok
- Department of Clinical Pharmacology, Kings College London, St Thomas' Hospital, London, UK
| | - Lorraine Bruce
- British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Nicholas L Mills
- British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK; Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Matthew A Bailey
- British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - David J Webb
- British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK; Clinical Research Centre, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - James W Dear
- British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Neeraj Dhaun
- British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK; Clinical Research Centre, University of Edinburgh, Western General Hospital, Edinburgh, UK; Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, UK.
| |
Collapse
|
10
|
Monocytes are increased in pregnancy after gestational hypertensive disease. Sci Rep 2022; 12:10358. [PMID: 35725746 PMCID: PMC9209470 DOI: 10.1038/s41598-022-13606-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 05/25/2022] [Indexed: 11/08/2022] Open
Abstract
Monocytes derive from bone marrow and circulate in the blood. They phagocytose, produce cytokines and present antigens. Individual monocyte subsets play distinct roles in the pathogenesis of cardiovascular disease, but their implications in gestational hypertensive disease are unclear. Our objective was to examine the difference in monocyte subsets between pregnant women with or without previous hypertension in pregnancy. Women were enrolled in a prospective observational study in which monoclonal antibodies against cell surface receptors were used to detect monocytes in the peripheral blood by flow cytometry. We compared 17 pregnant women with previous hypertension in pregnancy (Group 1) and 42 pregnant women without previous gestational hypertensive disease (Group 2) with 27 healthy, non-pregnant controls (Group 3). The pregnant women were studied at 13 ± 1 weeks gestation. Monocyte subsets were quantified by flow cytometry: Mon1 (CD14++CD16-CCR2+), Mon2 (CD14++CD16+CCR2+), Mon3 (CD14+CD16+CCR2-), their aggregates with platelets and expression of the surface markers. The groups were well-matched for age, body mass index and ethnicity (P > 0.05 for all). Mon1 counts were higher in women with a history of gestational hypertension or preeclampsia compared to other groups (Group 1 = 441 per µl (376-512); Group 2 = 357 (309-457); Group 3 = 323 (277-397); P < 0.001). Mon3 was higher in both groups of pregnant women compared to non-pregnant controls (Group 1 = 51 (38-62); Group 2 = 38 (29-58); Group 3 = 26 (20-40), P = 0.002). Increased monocytes in women with a previous hypertensive pregnancy generates a hypothesis that these cells may link hypertension in pregnancy, chronic inflammation and future cardiovascular risk.
Collapse
|
11
|
Richardson A, Krivokhizhina T, Lorkiewicz P, D’Souza S, Bhatnagar A, Srivastava S, Conklin DJ. Effects of electronic cigarette flavorants on human platelet aggregation ex vivo. Toxicol Rep 2022; 9:814-820. [PMID: 36518374 PMCID: PMC9742839 DOI: 10.1016/j.toxrep.2022.04.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 01/11/2023] Open
Abstract
Because little is known about the effects of individual flavorants in electronic cigarette (e-cig) fluids on human platelet aggregation, we tested for the direct effects of 15 common e-cig flavorants on adenosine diphosphate (ADP)-induced human platelet aggregation ex vivo. To better understand a potential mechanism of action of flavorants, we quantified 2 phases of aggregation. Human platelet-rich plasma (PRP) was obtained from whole blood of healthy volunteers and used in a platelet aggregometry assay. PRP was incubated with 1 of 15 different flavorant compounds (e.g., benzyl alcohol, eugenol, citronellol, menthol, menthone, diacetyl, maltol, limonene, methylbutyric acid, isoamyl acetate, acetylpyridine, eucalyptol, 2,5-dimethylpyrazine, cinnamaldehyde, and vanillin) at 100 µM for 5 min at 37 °C prior to addition of ADP (10 µM). Subsequent ADP-induced platelet aggregation was tracked for 5 min using an aggregometer. Aggregation curves were analyzed for flavorant-induced effects on total (%) aggregation, Phase 1 and Phase 2 components, and compared with their ADP-only control via One-Way ANOVA. Notably, eugenol significantly inhibited total aggregation; an effect due solely to inhibition of Phase 2. No other flavor tested had any effect on total or phase-specific ADP-induced platelet aggregation. These results indicate that parent flavorant compounds commonly found in e-cig liquids neither activate nor inhibit ADP-induced human platelet aggregation. However, as flavorants are chemically altered during heating of e-cig, thermally-derived products of flavorants (e.g., flavor acetals) also will need to be tested for effects on platelet activation.
Collapse
Key Words
- AA, arachidonic acid
- ADP, adenosine diphosphate
- CVD, cardiovascular disease
- E-cig, electronic cigarette
- ENDS,, electronic nicotine delivery systems
- Electronic cigarettes
- Electronic nicotine delivery systems
- Eugenol
- Flavorants
- GRAS, generally regarded as safe
- NO, nitric oxide
- P1,, phase 1 aggregation
- P2,, phase 2 aggregation
- PAF, platelet activating factor
- PG, propylene glycol
- PPP,, platelet-poor plasma
- PRP,, platelet-rich plasma
- ROS, reactive oxygen species
- Thrombosis
- Tobacco
- TxA2, thromboxane A2
- VG, vegetable glycerol
Collapse
Affiliation(s)
- Andre Richardson
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, USA
- Diabetes & Obesity Center, University of Louisville, USA
| | - Tatiana Krivokhizhina
- Christina Lee Brown Envirome Institute, University of Louisville, USA
- Diabetes & Obesity Center, University of Louisville, USA
| | - Pawel Lorkiewicz
- Christina Lee Brown Envirome Institute, University of Louisville, USA
- Diabetes & Obesity Center, University of Louisville, USA
- Department of Chemistry, University of Louisville, USA
| | - Stanley D’Souza
- Department of Physiology, School of Medicine, University of Louisville, USA
| | - Aruni Bhatnagar
- Christina Lee Brown Envirome Institute, University of Louisville, USA
- Diabetes & Obesity Center, University of Louisville, USA
- Division of Environmental Medicine, Department of Medicine, University of Louisville, USA
| | - Sanjay Srivastava
- Christina Lee Brown Envirome Institute, University of Louisville, USA
- Diabetes & Obesity Center, University of Louisville, USA
- Division of Environmental Medicine, Department of Medicine, University of Louisville, USA
| | - Daniel J. Conklin
- Christina Lee Brown Envirome Institute, University of Louisville, USA
- Diabetes & Obesity Center, University of Louisville, USA
- Division of Environmental Medicine, Department of Medicine, University of Louisville, USA
| |
Collapse
|
12
|
Yang S, Zheng X, Qian M, Wang H, Wang F, Wei Y, Midgley AC, He J, Tian H, Zhao Q. Nitrate-Functionalized poly(ε-Caprolactone) Small-Diameter Vascular Grafts Enhance Vascular Regeneration via Sustained Release of Nitric Oxide. Front Bioeng Biotechnol 2021; 9:770121. [PMID: 34917597 PMCID: PMC8670382 DOI: 10.3389/fbioe.2021.770121] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/04/2021] [Indexed: 01/04/2023] Open
Abstract
Artificial small-diameter vascular grafts (SDVG) fabricated from synthetic biodegradable polymers, such as poly(ε-caprolactone) (PCL), exhibit beneficial mechanical properties but are often faced with issues impacting their long-term graft success. Nitric oxide (NO) is an important physiological gasotransmitter with multiple roles in orchestrating vascular tissue function and regeneration. We fabricated a functional vascular graft by electrospinning of nitrate-functionalized poly(ε-caprolactone) that could release NO in a sustained manner via stepwise biotransformation in vivo. Nitrate-functionalized SDVG (PCL/NO) maintained patency following abdominal arterial replacement in rats. PCL/NO promoted cell infiltration at 3-months post-transplantation. In contrast, unmodified PCL SDVG showed slow cell in-growth and increased incidence of neointima formation. PCL/NO demonstrated improved endothelial cell (EC) alignment and luminal coverage, and more defined vascular smooth muscle cell (VSMC) layer, compared to unmodified PCL SDVG. In addition, release of NO stimulated Sca-1+ vascular progenitor cells (VPCs) to differentiate and contribute to rapid luminal endothelialization. Furthermore, PCL/NO inhibited the differentiation of VPCs into osteopontin-positive cells, thereby preventing vascular calcification. Overall, PCL/NO demonstrated enhanced cell ingrowth, EC monolayer formation and VSMC layer regeneration; whilst inhibiting calcified plaque formation. Our results suggested that PCL/NO could serve as promising candidates for improved and long-term success of SDVG implants.
Collapse
Affiliation(s)
- Sen Yang
- Department of Peripheral Vascular Disease, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Department of Vascular Surgery, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Xueni Zheng
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China.,Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, China
| | - Meng Qian
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China.,Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, China
| | - He Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China.,Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, China
| | - Fei Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China.,Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, China
| | - Yongzhen Wei
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China.,Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, China
| | - Adam C Midgley
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China.,Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, China
| | - Ju He
- Department of Vascular Surgery, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Hongyan Tian
- Department of Peripheral Vascular Disease, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qiang Zhao
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China.,Key Laboratory of Bioactive Materials (Ministry of Education), College of Life Sciences, Nankai University, Tianjin, China.,Zhengzhou Cardiovascular Hospital and 7th People's Hospital of Zhengzhou, Zhengzhou, China
| |
Collapse
|
13
|
Johny E, Bhaskar P, Alam MJ, Kuladhipati I, Das R, Adela R. Platelet Mediated Inflammation in Coronary Artery Disease with Type 2 Diabetes Patients. J Inflamm Res 2021; 14:5131-5147. [PMID: 34675593 PMCID: PMC8504552 DOI: 10.2147/jir.s326716] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/19/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is a well-established risk factor for the development of atherosclerotic coronary artery disease. Platelet hyperactivity and inflammation are associated with the development of coronary artery disease (CAD) in T2DM patients. We investigated the status of immune cells, platelet activation, and platelet-immune cell interactions in T2DM_CAD patients. METHODOLOGY The study population consisted of four groups of subjects, healthy control (CT, n = 20), T2DM (n = 44), CAD (n = 20) and T2DM_CAD (n = 38). Platelet activation, immunome profiling and platelet-immune cell interactions were analysed by flow cytometry. The circulatory levels of inflammatory cytokines/chemokines were assessed using multiplex assay. RESULTS Increased platelet activation and increased platelet-immune cell aggregate formation were observed in T2DM and T2DM_CAD groups compared to the control and CAD groups (p < 0.05). Our immunome profile analysis revealed, altered monocyte subpopulations and dendritic cell populations in T2DM, CAD and T2DM_CAD groups compared to the control group (p < 0.05). Furthermore, significantly increased IL-1β, IL-2, IL-4, IL-6, IL-8, IL12p70, IL-13 IL-18, CCL2, and decreased CXCL1, CCL5 levels were observed in T2DM_CAD group compared to the control group. Our ex-vivo study increased platelet-monocyte aggregate formation was observed upon D-glucose exposure in a time and concentration dependent manner. CONCLUSION Our data suggests that T2DM, CAD and T2DM_CAD are associated with altered immune cell populations. Furthermore, it has been confirmed that hyperglycemia induces platelet activation and forms platelet-immune cell aggregation which may lead to the release of inflammatory cytokines and chemokines and contribute to the complexity of CAD and type 2 diabetes.
Collapse
Affiliation(s)
- Ebin Johny
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Guwahati, Assam, 781101, India
| | - Pathoori Bhaskar
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Guwahati, Assam, 781101, India
| | - Md Jahangir Alam
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Guwahati, Assam, 781101, India
| | | | - Rupam Das
- Down Town Hospital, Guwahati, Assam, 781006, India
| | - Ramu Adela
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Guwahati, Assam, 781101, India
| |
Collapse
|
14
|
Oggero S, de Gaetano M, Marcone S, Fitzsimons S, Pinto AL, Ikramova D, Barry M, Burke D, Montero-Melendez T, Cooper D, Burgoyne T, Belton O, Norling LV, Brennan EP, Godson C, Perretti M. Extracellular vesicles from monocyte/platelet aggregates modulate human atherosclerotic plaque reactivity. J Extracell Vesicles 2021; 10:12084. [PMID: 33936566 PMCID: PMC8077084 DOI: 10.1002/jev2.12084] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 03/24/2021] [Accepted: 04/01/2021] [Indexed: 12/16/2022] Open
Abstract
Extracellular vesicles (EVs) are emerging as key players in different stages of atherosclerosis. Here we provide evidence that EVs released by mixed aggregates of monocytes and platelets in response to TNF‐α display pro‐inflammatory actions on endothelial cells and atherosclerotic plaques. Tempering platelet activation with Iloprost, Aspirin or a P2Y12 inhibitor impacted quantity and phenotype of EV produced. Proteomics of EVs from cells activated with TNF‐α alone or in the presence of Iloprost revealed a distinct composition, with interesting hits like annexin‐A1 and gelsolin. When added to human atherosclerotic plaque explants, EVs from TNF‐α stimulated monocytes augmented release of cytokines. In contrast, EVs generated by TNF‐α together with Iloprost produced minimal plaque activation. Notably, patients with coronary artery disease that required percutaneous coronary intervention had elevated plasma numbers of monocyte, platelet as well as double positive EV subsets. In conclusion, EVs released following monocyte/platelet activation may play a potential role in the development and progression of atherosclerosis. Whereas attenuating platelet activation modifies EV composition released from monocyte/platelet aggregates, curbing their pro‐inflammatory actions may offer therapeutic avenues for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Silvia Oggero
- William Harvey Research Institute Bart's and the London School of Medicine Queen Mary University of London London UK
| | - Monica de Gaetano
- Diabetes Complications Research Centre Conway Institute, & School of Medicine University College Dublin Dublin Ireland
| | - Simone Marcone
- Trinity Translational Medicine Institute Trinity College Dublin Dublin Ireland
| | - Stephen Fitzsimons
- Diabetes Complications Research Centre Conway Institute, & School of Medicine University College Dublin Dublin Ireland
| | | | - Dinara Ikramova
- School of Engineering and Materials Science Queen Mary University of London London UK
| | - Mary Barry
- Department of Vascular Surgery St. Vincent's University Hospital Dublin Ireland
| | - David Burke
- Department of Vascular Surgery St. Vincent's University Hospital Dublin Ireland
| | - Trinidad Montero-Melendez
- William Harvey Research Institute Bart's and the London School of Medicine Queen Mary University of London London UK.,Centre for inflammation and Therapeutic Innovation Queen Mary University of London London UK
| | - Dianne Cooper
- William Harvey Research Institute Bart's and the London School of Medicine Queen Mary University of London London UK.,Centre for inflammation and Therapeutic Innovation Queen Mary University of London London UK
| | - Thomas Burgoyne
- Royal Brompton & Harefield NHS Foundation Trust London UK.,Institute of Ophthalmology, Faculty of Brain Sciences University College London London UK
| | - Orina Belton
- Diabetes Complications Research Centre Conway Institute, & School of Medicine University College Dublin Dublin Ireland
| | - Lucy V Norling
- William Harvey Research Institute Bart's and the London School of Medicine Queen Mary University of London London UK.,Centre for inflammation and Therapeutic Innovation Queen Mary University of London London UK
| | - Eoin P Brennan
- Diabetes Complications Research Centre Conway Institute, & School of Medicine University College Dublin Dublin Ireland
| | - Catherine Godson
- Diabetes Complications Research Centre Conway Institute, & School of Medicine University College Dublin Dublin Ireland
| | - Mauro Perretti
- William Harvey Research Institute Bart's and the London School of Medicine Queen Mary University of London London UK.,Centre for inflammation and Therapeutic Innovation Queen Mary University of London London UK
| |
Collapse
|
15
|
A study of endothelial and platelet microvesicles across different hypertension phenotypes. J Hum Hypertens 2021; 36:561-569. [PMID: 33837293 DOI: 10.1038/s41371-021-00531-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/05/2021] [Accepted: 03/22/2021] [Indexed: 01/01/2023]
Abstract
Rather than being mere biomarkers reflecting generalized vascular injury, endothelial- (EMVs) and platelet-derived (PMVs) microvesicles have emerged as potent regulators of intercellular communication with significant biologic effects in vascular homeostasis and several pathophysiological responses including inflammation and thrombosis. So far, studies in hypertension are scarce, whereas no studies exist in masked hypertension (MH). We measured EMVs and PMVs in untreated, newly diagnosed hypertensives (HTs) and MHs compared to normotensive controls (NTs), and associated them with various cardiovascular risk factors. Sustained hypertension (SHT) and MH were defined according to standard blood pressure (BP) criteria. All HTs were free of cardiovascular disease and medications. Microvesicles' quantitation and detection were performed by flow cytometry by using cell-specific antibodies and corresponding isotypes (anti-CD105 and anti-CD144 for EMVs, anti-CD42a for PMVs, and Annexin V-fluorescein isothiocyanate for all microvesicles). In this study, we included 59 HTs (44 SHTs and 15 MHs) and 27 NTs. HTs had significantly elevated EMVs (p = 0.004), but not PMVs compared to NTs. MHs had significantly elevated EMVs compared to NTs (p = 0.012) but not compared to SHTs. Furthermore, EMVs significantly correlated with ambulatory (r = 0.214-0.284), central BP (r = 0.247-0.262), and total vascular resistance (r = 0.327-0.361). EMVs are increased not only in SHTs but also in MHs, a hypertension phenotype with a cardiovascular risk close to SHT. EMVs have emerged as active contributors to thromboinflammation and vascular damage and may explain, in part, the adverse cardiovascular profile of SHTs and MHs.
Collapse
|
16
|
Mohtashami M, Razavi A, Abolhassani H, Aghamohammadi A, Yazdani R. Primary Immunodeficiency and Thrombocytopenia. Int Rev Immunol 2021; 41:135-159. [PMID: 33464134 DOI: 10.1080/08830185.2020.1868454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Primary immunodeficiency (PID) or Inborn errors of immunity (IEI) refers to a heterogeneous group of disorders characterized by immune system impairment. Although patients with IEI manifest highly variable symptoms, the most common clinical manifestations are recurrent infections, autoimmunity and malignancies. Some patients present hematological abnormality including thrombocytopenia due to different pathogenic mechanisms. This review focuses on primary and secondary thrombocytopenia as a complication, which can occur in IEI. Based on the International Union of Immunological Societies phenotypic classification for IEI, the several innate and adaptive immunodeficiency disorders can lead to thrombocytopenia. This review, for the first time, describes manifestation, mechanism and therapeutic modalities for thrombocytopenia in different classes of IEI.
Collapse
Affiliation(s)
- Maryam Mohtashami
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran.,Research Center for Immunodeficiencies, Tehran University of Medical Sciences, Tehran, Iran
| | - Azadehsadat Razavi
- Research Center for Immunodeficiencies, Tehran University of Medical Sciences, Tehran, Iran.,Department of Animal Biology, Faculty of Biology Sciences, University of Kharazmi, Tehran, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hassan Abolhassani
- Research Center for Immunodeficiencies, Tehran University of Medical Sciences, Tehran, Iran.,Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Asghar Aghamohammadi
- Research Center for Immunodeficiencies, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Yazdani
- Research Center for Immunodeficiencies, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Yaribeygi H, Atkin SL, Jamialahmadi T, Sahebkar A. A Review on the Effects of New Anti-Diabetic Drugs on Platelet Function. Endocr Metab Immune Disord Drug Targets 2021; 20:328-334. [PMID: 31612835 DOI: 10.2174/1871530319666191014110414] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 08/05/2019] [Accepted: 09/13/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Cardiovascular complications account for the majority of deaths caused by diabetes mellitus. Platelet hyperactivity has been shown to increase the risk of thrombotic events and is a therapeutic target for their prevention in diabetes. Modulation of platelet function by diabetes agents in addition to their hypoglycemic effects would contribute to cardiovascular protection. Newly introduced antidiabetic drugs of sodium-glucose cotransporter 2 inhibitors (SGLT2i), glucagon like peptide-1 receptor agonists (GLP-1RA) and dipeptidyl peptidase-4 inhibitors may have anti-platelet effects, and in the case of SGLT2i and GLP-1RA may contribute to their proven cardiovascular benefit that has been shown clinically. OBJECTIVE Here, we reviewed the potential effects of these agents on platelet function in diabetes. RESULTS AND CONCLUSION GLP-1RA and DPP-4i drugs have antiplatelet properties beyond their primary hypoglycemic effects. Whilst we have little direct evidence for the antiplatelet effects of SGLT2 inhibitors, some studies have shown that these agents may inhibit platelet aggregation and reduce the risk of thrombotic events in diabetes.
Collapse
Affiliation(s)
- Habib Yaribeygi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Tannaz Jamialahmadi
- Halal Research Center of IRI, FDA, Tehran, Iran.,Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
18
|
Assessment of Endothelial Injury and Pro-Coagulant Activity Using Circulating Microvesicles in Survivors of Allogeneic Hematopoietic Cell Transplantation. Int J Mol Sci 2020; 21:ijms21249768. [PMID: 33371421 PMCID: PMC7767425 DOI: 10.3390/ijms21249768] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 02/07/2023] Open
Abstract
(1) Background: survivors of allogeneic hematopoietic cell transplantation (alloHCT) suffer from morbidity and mortality due to cardiovascular events. We hypothesized that vascular injury and pro-coagulant activity are evident in alloHCT survivors without existing alloHCT complications or relapse. (2) Methods: we enrolled consecutive adult alloHCT survivors without established cardiovascular disease and control individuals matched for traditional cardiovascular risk factors (January-December 2019). Circulating microvesicles (MVs) of different cellular origins (platelet, erythrocyte, and endothelial) were measured by a standardized flow cytometry protocol as novel markers of vascular injury and pro-coagulant activity. (3) Results: we recruited 45 survivors after a median of 2.3 (range 1.1-13.2) years from alloHCT, and 45 controls. The majority of patients suffered from acute (44%) and/or chronic (66%) graft-versus-host disease (GVHD). Although the two groups were matched for traditional cardiovascular risk factors, alloHCT survivors showed significantly increased platelet and erythrocyte MVs compared to controls. Within alloHCT survivors, erythrocyte MVs were significantly increased in patients with a previous history of thrombotic microangiopathy. Interestingly, endothelial MVs were significantly increased only in alloHCT recipients of a myeloablative conditioning. Furthermore, MVs of different origins showed a positive association with each other. (4) Conclusions: endothelial dysfunction and increased thrombotic risk are evident in alloHCT recipients long after alloHCT, independently of traditional cardiovascular risk factors. An apparent synergism of these pathophysiological processes may be strongly involved in the subsequent establishment of cardiovascular disease.
Collapse
|
19
|
Chimen M, Evryviadou A, Box CL, Harrison MJ, Hazeldine J, Dib LH, Kuravi SJ, Payne H, Price JMJ, Kavanagh D, Iqbal AJ, Lax S, Kalia N, Brill A, Thomas SG, Belli A, Crombie N, Adams RA, Evans SA, Deckmyn H, Lord JM, Harrison P, Watson SP, Nash GB, Rainger GE. Appropriation of GPIbα from platelet-derived extracellular vesicles supports monocyte recruitment in systemic inflammation. Haematologica 2020; 105:1248-1261. [PMID: 31467123 PMCID: PMC7193470 DOI: 10.3324/haematol.2018.215145] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 08/23/2019] [Indexed: 01/13/2023] Open
Abstract
Interactions between platelets, leukocytes and the vessel wall provide alternative pathological routes of thrombo-inflammatory leukocyte recruitment. We found that when platelets were activated by a range of agonists in whole blood, they shed platelet-derived extracellular vesicles which rapidly and preferentially bound to blood monocytes compared to other leukocytes. Platelet-derived extracellular vesicle binding to monocytes was initiated by P-selectin-dependent adhesion and was stabilised by binding of phosphatidylserine. These interactions resulted in the progressive transfer of the platelet adhesion receptor GPIbα to monocytes. GPIbα+-monocytes tethered and rolled on immobilised von Willebrand Factor or were recruited and activated on endothelial cells treated with TGF-β1 to induce the expression of von Willebrand Factor. In both models monocyte adhesion was ablated by a function-blocking antibody against GPIbα. Monocytes could also bind platelet-derived extracellular vesicle in mouse blood in vitro and in vivo Intratracheal instillations of diesel nanoparticles, to model chronic pulmonary inflammation, induced accumulation of GPIbα on circulating monocytes. In intravital experiments, GPIbα+-monocytes adhered to the microcirculation of the TGF-β1-stimulated cremaster muscle, while in the ApoE-/- model of atherosclerosis, GPIbα+-monocytes adhered to the carotid arteries. In trauma patients, monocytes bore platelet markers within 1 hour of injury, the levels of which correlated with severity of trauma and resulted in monocyte clearance from the circulation. Thus, we have defined a novel thrombo-inflammatory pathway in which platelet-derived extracellular vesicles transfer a platelet adhesion receptor to monocytes, allowing their recruitment in large and small blood vessels, and which is likely to be pathogenic.
Collapse
Affiliation(s)
- Myriam Chimen
- Institute of Cardiovascular Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham, UK
- NIHR Surgical Reconstruction and Microbiology Research Centre, Institute of Inflammation and Ageing, Birmingham University Medical School, Birmingham, UK
| | - Aigli Evryviadou
- Institute of Cardiovascular Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham, UK
| | - Clare L Box
- Institute of Cardiovascular Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham, UK
| | - Matthew J Harrison
- Institute of Cardiovascular Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham, UK
| | - Jon Hazeldine
- NIHR Surgical Reconstruction and Microbiology Research Centre, Institute of Inflammation and Ageing, Birmingham University Medical School, Birmingham, UK
| | - Lea H Dib
- Institute of Cardiovascular Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham, UK
| | - Sahithi J Kuravi
- Institute of Cardiovascular Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham, UK
| | - Holly Payne
- Institute of Cardiovascular Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham, UK
| | - Joshua M J Price
- NIHR Surgical Reconstruction and Microbiology Research Centre, Institute of Inflammation and Ageing, Birmingham University Medical School, Birmingham, UK
| | - Dean Kavanagh
- Institute of Cardiovascular Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham, UK
| | - Asif J Iqbal
- Institute of Cardiovascular Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham, UK
| | - Sian Lax
- Institute of Cardiovascular Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham, UK
| | - Neena Kalia
- Institute of Cardiovascular Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham, UK
| | - Alex Brill
- Institute of Cardiovascular Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham and Nottingham, The Midlands, UK
- Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Steve G Thomas
- Institute of Cardiovascular Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham and Nottingham, The Midlands, UK
| | - Antonio Belli
- NIHR Surgical Reconstruction and Microbiology Research Centre, Institute of Inflammation and Ageing, Birmingham University Medical School, Birmingham, UK
| | - Nicholas Crombie
- NIHR Surgical Reconstruction and Microbiology Research Centre, Institute of Inflammation and Ageing, Birmingham University Medical School, Birmingham, UK
| | - Rachel A Adams
- Cardiff School of Health Sciences, Cardiff Metropolitan University, Cardiff, UK
| | - Shelley-Ann Evans
- Cardiff School of Health Sciences, Cardiff Metropolitan University, Cardiff, UK
| | - Hans Deckmyn
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Janet M Lord
- NIHR Surgical Reconstruction and Microbiology Research Centre, Institute of Inflammation and Ageing, Birmingham University Medical School, Birmingham, UK
| | - Paul Harrison
- NIHR Surgical Reconstruction and Microbiology Research Centre, Institute of Inflammation and Ageing, Birmingham University Medical School, Birmingham, UK
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham and Nottingham, The Midlands, UK
| | - Gerard B Nash
- Institute of Cardiovascular Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham, UK
| | - G Ed Rainger
- Institute of Cardiovascular Sciences, College of Medicine and Dentistry, University of Birmingham, Birmingham, UK
| |
Collapse
|
20
|
An X, Jiang G, Cheng C, Lv Z, Liu Y, Wang F. Inhibition of Platelets by Clopidogrel Suppressed Ang II-Induced Vascular Inflammation, Oxidative Stress, and Remodeling. J Am Heart Assoc 2019; 7:e009600. [PMID: 30608200 PMCID: PMC6404205 DOI: 10.1161/jaha.118.009600] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Platelets play a role in promoting inflammatory responses under several disease conditions. Platelets are activated in hypertensive patients. However, the mechanisms responsible for platelet‐mediating vascular inflammation are unknown. The present study investigated the role of platelets in promoting vascular inflammation following angiotensin II (Ang II) stimulation, and the efficacy of antiplatelet intervention. Methods and Results Within a mouse model of Ang II infusion (490 ng/kg per min), we measured the portion of P‐selectin–positive platelets and platelet‐monocyte (P‐M) binding in blood samples, and platelet accumulation and P‐M binding in vessels under Ang II stimulation at days 1, 3, and 7. We tested the efficacy of clopidogrel (15 mg/kg per day, followed by 5 mg/kg per day) on Ang II‐induced platelet activation, P‐M binding, vascular platelet accumulation, as well as vascular inflammation and remodeling at day 7 or 14. Clopidogrel reduced platelet vascular deposition (28.7±2.4% versus 18.3±2.9%), suppressed inflammatory cell infiltration (3.6±0.8×104/vessel versus 2.3±1.2×104/vessel) and oxidative stress, and attenuated vascular remodeling and dysfunction (55.0±5.5% versus 84.0±6.0%) following Ang II stimulation at day 7 or 14. Clopidogrel suppressed Ang II‐induced P‐M binding both at circulating (13.4±3.3% versus 5.9±2.7%) and regional (33.4±4.3% versus 11.9±2.7%) levels. Conclusions Platelets play a critical role in vascular inflammation under Ang II stimulation, with a marked promotion of P‐M binding as an important mechanism. Clopidogrel prevented vascular inflammation in Ang II‐infused mice.
Collapse
Affiliation(s)
- Xiangbo An
- 1 Department of Interventional Therapy the First Affiliated Hospital of Dalian Medical University Dalian China.,3 Institute of Cardiovascular Diseases the First Affiliated Hospital of Dalian Medical University Dalian China
| | - Guinan Jiang
- 1 Department of Interventional Therapy the First Affiliated Hospital of Dalian Medical University Dalian China.,3 Institute of Cardiovascular Diseases the First Affiliated Hospital of Dalian Medical University Dalian China
| | - Cheng Cheng
- 4 Center for Clinical Research on Neurological Diseases the First Affiliated Hospital of Dalian Medical University Dalian China
| | - Zhengshuai Lv
- 2 Department of Anesthesia the First Affiliated Hospital of Dalian Medical University Dalian China
| | - Yang Liu
- 3 Institute of Cardiovascular Diseases the First Affiliated Hospital of Dalian Medical University Dalian China
| | - Feng Wang
- 1 Department of Interventional Therapy the First Affiliated Hospital of Dalian Medical University Dalian China
| |
Collapse
|
21
|
Gavriilaki E, Gkaliagkousi E, Sakellari I, Anyfanti P, Douma S, Anagnostopoulos A. Early Prediction of Cardiovascular Risk after Hematopoietic Cell Transplantation: Are We There Yet? Biol Blood Marrow Transplant 2019; 25:e310-e316. [PMID: 31310812 DOI: 10.1016/j.bbmt.2019.07.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 07/03/2019] [Accepted: 07/10/2019] [Indexed: 12/14/2022]
Abstract
Cardiovascular (CV) events have emerged as a major cause of morbidity and mortality among hematopoietic cell transplantation (HCT) survivors. Accumulating evidence supports the presence of increased CV risk in HCT recipients. Most studies have focused mainly on traditional CV risk factors, such as the metabolic syndrome and hypertension. However, detection of these factors suggests the development of irreversible overt clinical atherosclerosis. Therefore, earlier prediction of CV risk is needed to prevent CV morbidity and mortality in these patients. In the field of CV research, endothelial dysfunction is considered an early event in the pathophysiology of CV risk factors, and a number of markers have been proposed for its assessment. In addition, markers of subclinical target organ damage have been introduced to implement CV risk prediction and early preventive or intensive therapeutic interventions. Furthermore, a number of CV models have been suggested aiming for optimal stratification of patients. Preliminary studies have indicated excess CV risk using these early markers in HCT recipients. However, their role in the pathophysiology and clinical practice in HCT survivors remains largely understudied. Taking into account the need for increased awareness from treating physicians in this evolving setting, we conducted a state-of-the-art review aiming to summarize current knowledge on endothelial dysfunction, subclinical target organ damage, and CV risk prediction in HCT survivors.
Collapse
Affiliation(s)
- Eleni Gavriilaki
- Hematology Department-BMT Unit, G. Papanicolaou Hospital, Thessaloniki, Greece.
| | - Eugenia Gkaliagkousi
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioanna Sakellari
- Hematology Department-BMT Unit, G. Papanicolaou Hospital, Thessaloniki, Greece
| | - Panagiota Anyfanti
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Stella Douma
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | |
Collapse
|
22
|
Papain Ameliorates the MPAs Formation-Mediated Activation of Monocytes by Inhibiting Cox-2 Expression via Regulating the MAPKs and PI3K/Akt Signal Pathway. BIOMED RESEARCH INTERNATIONAL 2018; 2018:3632084. [PMID: 30410927 PMCID: PMC6206584 DOI: 10.1155/2018/3632084] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 08/29/2018] [Accepted: 09/25/2018] [Indexed: 12/30/2022]
Abstract
Monocytes activation and subsequent inflammatory response mediated by monocyte-platelet aggregates (MPAs) formation play the key roles in the early pathogenesis of atherosclerosis (AS). Exploration of novel drugs to ameliorate MPAs formation-mediated monocytes activation would be helpful for the treatment of AS patients. Papain has definite pharmacological effects including antiplatelet, thrombolysis, and anti-inflammation. However, its effect on MPAs formation and the following monocytes activation remains vague. This study aimed to illustrate the underlying mechanisms of papain on MPAs formation-initiated monocytes activation in vitro. In this study, Papain, Cox-2 inhibitor (NS-398), and NF-κB agonist (TNF-α) were used as the treating agents, respectively. MPAs formation and activated monocytes were measured by flow cytometry (FCM). Cox-2 mRNA, MCP-1, and proteins of Cox-2 and NF-κB signal pathway were detected by qRT-PCR, ELISA, and western blotting, respectively. As we observed, papain exhibited the powerful inhibitory effects on thrombin-mediated MPAs formation and monocytes activation in a concentration-dependent manner as what Cox-2 inhibitor demonstrated. However, the inhibitory tendency was significantly reversed by TNF-α. We also discovered that both Cox-2 mRNA and protein expression as well as the release of MCP-1 of monocyte was inhibited by either papain or NS-398, but TNF-α stimulated Cox-2 expression and release of MCP-1. The results of western blotting assay indicated that thrombin-mediated proteins expression of MAPKs and PI3K/Akt signal pathway was inhibited by papain and NS-398. However, TNF-α notably abated the inhibitory effects of papain on the process of MPAs-initiated monocytes activation. Our findings suggest that papain can inhibit the MPAs formation-mediated activation of monocytes by inhibiting the MAPKs and PI3K/Akt signal pathway.
Collapse
|
23
|
Robson R, Kundur AR, Singh I. Oxidative stress biomarkers in type 2 diabetes mellitus for assessment of cardiovascular disease risk. Diabetes Metab Syndr 2018; 12:455-462. [PMID: 29307576 DOI: 10.1016/j.dsx.2017.12.029] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 12/27/2017] [Indexed: 01/07/2023]
Abstract
AIMS Type-2 Diabetes Mellitus (T2DM) is one of the most prevalent and progressive metabolic conditions affecting approximately 8.5% of the global population. Individuals with T2DM have a significantly increased risk of developing chronic conditions such as cardiovascular disease (CVD) and its associated complications, therefore, it is of great importance to establish strategies for combatting T2DM and its associated chronic conditions. Current literature has identified several biomarkers that are known to play a key role in the pathogenesis of CVD. Many of these biomarkers affecting CVD are influenced by an increase in oxidative stress as seen in T2DM. The purpose of this review is to analyse and correlate the oxidative stress-related biomarkers that have been identified in the literature to provide an updated summary of their significance in CVD risk factors. DATA SYNTHESIS This review has analysed current research on T2DM, CVD, and oxidative stress. Four key cardiovascular risk factors: thrombosis, inflammation, vascular homeostasis and cellular proliferation were searched to identify potential biomarkers for this review. These biomarkers stem from seven major cellular pathways; NF-κB, Keap1-Nrf2, protein kinase-C, macrophage activation, arachidonic acid mobilisation, endothelial dysfunction and advanced glycation end products. CONCLUSIONS The pathways and biomarkers were analysed to show their role as contributing factors to CVD development and a summary is made regarding the assessment of cardiovascular risk in T2DM individuals.
Collapse
Affiliation(s)
- Roy Robson
- School of Medical Science, Menzies Health Institute Queensland, Gold Coast Campus, Griffith University, QLD 4222, Australia
| | - Avinash R Kundur
- School of Medical Science, Menzies Health Institute Queensland, Gold Coast Campus, Griffith University, QLD 4222, Australia
| | - Indu Singh
- School of Medical Science, Menzies Health Institute Queensland, Gold Coast Campus, Griffith University, QLD 4222, Australia.
| |
Collapse
|
24
|
Gkaliagkousi E, Gavriilaki E, Triantafyllou A, Nikolaidou B, Anyfanti P, Koletsos N, Vamvakis A, Dipla K, Lazaridis A, Douma S. Asymmetric dimethylarginine levels are associated with augmentation index across naïve untreated patients with different hypertension phenotypes. J Clin Hypertens (Greenwich) 2018; 20:680-685. [DOI: 10.1111/jch.13237] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 01/08/2018] [Accepted: 01/12/2018] [Indexed: 01/11/2023]
Affiliation(s)
- Eugenia Gkaliagkousi
- 3rd Department of Internal Medicine; Papageorgiou Hospital; Aristotle University of Thessaloniki; Thessaloniki Greece
| | - Eleni Gavriilaki
- 3rd Department of Internal Medicine; Papageorgiou Hospital; Aristotle University of Thessaloniki; Thessaloniki Greece
| | - Areti Triantafyllou
- 3rd Department of Internal Medicine; Papageorgiou Hospital; Aristotle University of Thessaloniki; Thessaloniki Greece
| | - Barbara Nikolaidou
- 3rd Department of Internal Medicine; Papageorgiou Hospital; Aristotle University of Thessaloniki; Thessaloniki Greece
| | - Panagiota Anyfanti
- 3rd Department of Internal Medicine; Papageorgiou Hospital; Aristotle University of Thessaloniki; Thessaloniki Greece
| | - Nikolaos Koletsos
- 3rd Department of Internal Medicine; Papageorgiou Hospital; Aristotle University of Thessaloniki; Thessaloniki Greece
| | - Anastasios Vamvakis
- 3rd Department of Internal Medicine; Papageorgiou Hospital; Aristotle University of Thessaloniki; Thessaloniki Greece
| | - Konstantina Dipla
- Laboratory of Exercise Physiology and Biochemistry; Department of Physical Education and Sports Science; Aristotle University of Thessaloniki; Serres Greece
| | - Antonios Lazaridis
- 3rd Department of Internal Medicine; Papageorgiou Hospital; Aristotle University of Thessaloniki; Thessaloniki Greece
| | - Stella Douma
- 3rd Department of Internal Medicine; Papageorgiou Hospital; Aristotle University of Thessaloniki; Thessaloniki Greece
| |
Collapse
|
25
|
Extracellular Matrix Metalloproteinase Inducer EMMPRIN (CD147) in Cardiovascular Disease. Int J Mol Sci 2018; 19:ijms19020507. [PMID: 29419744 PMCID: PMC5855729 DOI: 10.3390/ijms19020507] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 01/31/2018] [Accepted: 02/01/2018] [Indexed: 12/22/2022] Open
Abstract
The receptor EMMPRIN is involved in the development and progression of cardiovascular diseases and in the pathogenesis of myocardial infarction. There are several binding partners of EMMPRIN mediating the effects of EMMPRIN in cardiovascular diseases. EMMPRIN interaction with most binding partners leads to disease progression by mediating cytokine or chemokine release, the activation of platelets and monocytes, as well as the formation of monocyte-platelet aggregates (MPAs). EMMPRIN is also involved in atherosclerosis by mediating the infiltration of pro-inflammatory cells. There is also evidence that EMMPRIN controls energy metabolism of cells and that EMMPRIN binding partners modulate intracellular glycosylation and trafficking of EMMPRIN towards the cell membrane. In this review, we systematically discuss these multifaceted roles of EMMPRIN and its interaction partners, such as Cyclophilins, in cardiovascular disease.
Collapse
|
26
|
Thrombogenicity and central pulse pressure to enhance prediction of ischemic event occurrence in patients with established coronary artery disease: The MAGMA-ischemia score. Atherosclerosis 2017; 268:55-62. [PMID: 29175655 DOI: 10.1016/j.atherosclerosis.2017.11.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 10/20/2017] [Accepted: 11/16/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIMS Conventional cardiovascular risk estimators based on clinical demographics have limited prediction of coronary events. Markers for thrombogenicity and vascular function have not been explored in risk estimation of high-risk patients with coronary artery disease. We aimed to develop a clinical and biomarker score to predict 3-year adverse cardiovascular events. METHODS Four hundred eleven patients, with ejection fraction ≥40% undergoing coronary angiography, and found to have a luminal diameter stenosis ≥50%, were included in the analysis. Thrombelastography indices and central pulse pressure (CPP) were determined at the time of catheterization. RESULTS We identified predictors of death, myocardial infarction (MI) or stroke and developed a numerical ischemia risk score. The primary endpoint of cardiovascular death, MI or stroke occurred in 22 patients (5.4%). The factors associated with events were age, prior PCI or CABG, diabetes, CPP, and thrombin-induced platelet-fibrin clot strength, and were included in the MAGMA-ischemia score. The MAGMA-ischemia score showed a c-statistic of 0.85 (95% Confidence Interval [CI] 0.80-0.87; p<0.001) for the primary endpoint. In the subset of patients who underwent revascularization, the c-statistic was 0.90 (p<0.001). Patients with MAGMA-ischemia score greater than 5 had highest risk to develop clinical events, hazard ratio for the primary endpoint: 13.9 (95% CI 5.8-33.1, p<0.001) and for the secondary endpoint: 4.8 (95% CI 2.3-9.6, p<0.001). When compared to previous models, the MAGMA-ischemia score yielded a higher discrimination. CONCLUSIONS Inclusion of CPP and assessment of thrombogenicity in a novel score for patients with documented CAD enhanced the prediction of events.
Collapse
|
27
|
Chen G, Bliden KP, Chaudhary R, Liu F, Kaza H, Navarese EP, Tantry US, Gurbel PA. Central aortic pulse pressure, thrombogenicity and cardiovascular risk. J Thromb Thrombolysis 2017; 44:223-233. [PMID: 28695310 DOI: 10.1007/s11239-017-1524-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
High central aortic pulse pressure (CPP) and thrombin-induced platelet-fibrin clot strength (TIP-FCS) have been associated with ischemic outcomes in patients with coronary artery disease in separate studies. But, the ischemic risk associated with these factors has never been analyzed in a single study and their interrelation is unknown. The primary aim of the study was to establish cut points for CPP and TIP-FCS measured at the time of catheterization associated with long term major adverse cardiovascular events. We enrolled 334 consecutive patients undergoing cardiac catheterization and assessed thrombogenicity by thrombelastography. Patients were followed up to 3 years. The primary endpoint was a composite of cardiovascular death, myocardial infarction, and ischemic stroke and the secondary endpoint was occurrence of the primary endpoint or recurrent ischemic events requiring hospitalization. Patients with primary and secondary endpoint occurrence had higher CPP (83 ± 20 vs. 60 ± 18 mmHg, p < 0.0001; 70 ± 21 vs. 59 ± 18 mmHg, p < 0.0001, respectively) and TIP-FCS (68.5 ± 5.8 vs. 65.5 ± 5.0 mm, p = 0.008; 67.4 ± 5.9 vs. 65.2 ± 4.8 mm, p = 0.001, respectively). CPP >60 mmHg and TIP-FCS >69 mm were both independent predictors of primary endpoint occurrence (p = 0.0001 and p = 0.02, respectively). ROC analysis for CPP and TIP-FCS showed a C-statistic of 0.81 (p < 0.0001) and 0.68 (p = 0.007) for the primary endpoint, respectively. Patients with CPP >60 mmHg had higher TIP-FCS (66.8 ± 5.1 vs. 64.8 ± 5.0 mm, p < 0.001) and primary and secondary endpoint occurrence (13 vs. 1.1%, p < 0.0001 and 31.8 vs. 14.4%, p = 0.0002, respectively). CPP >60 mmHg + TIP-FCS > 69 mm was associated with a markedly increased risk of primary endpoint occurrence [HR (95% CI) 5.4(2.3-12.5), p = 0.0001]. High CPP and thrombogenicity are interrelated; each are independently associated with increased cardiovascular risk; and simultaneous presence markedly enhances risk. The mechanistic link between CPP and thrombogenicity deserves further study.
Collapse
Affiliation(s)
- Gailing Chen
- Sinai Center for Thrombosis Research, Sinai Hospital, Baltimore, MD, USA.,Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Kevin P Bliden
- Inova Center for Thrombosis Research and Drug Development, Inova Heart and Vascular Institute, 3300 Gallows Rd, Fairfax, VA, USA
| | - Rahul Chaudhary
- Division of Medicine, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Fang Liu
- Sinai Center for Thrombosis Research, Sinai Hospital, Baltimore, MD, USA.,Department of Neurology, Beijing Hospital, Beijing, China
| | - Himabindu Kaza
- Sinai Center for Thrombosis Research, Sinai Hospital, Baltimore, MD, USA
| | - Eliano P Navarese
- Inova Center for Thrombosis Research and Drug Development, Inova Heart and Vascular Institute, 3300 Gallows Rd, Fairfax, VA, USA
| | - Udaya S Tantry
- Inova Center for Thrombosis Research and Drug Development, Inova Heart and Vascular Institute, 3300 Gallows Rd, Fairfax, VA, USA
| | - Paul A Gurbel
- Inova Center for Thrombosis Research and Drug Development, Inova Heart and Vascular Institute, 3300 Gallows Rd, Fairfax, VA, USA.
| |
Collapse
|
28
|
ÇEVİK Ö, ADIGÜZEL Z, BAYKAL AT, ŞENER A. Tumor necrosis factor-alpha induced caspase-3 activation-related iNOS gene expression in ADP-activated platelets. Turk J Biol 2017. [DOI: 10.3906/biy-1509-64] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
29
|
Zaldivia MTK, Rivera J, Hering D, Marusic P, Sata Y, Lim B, Eikelis N, Lee R, Lambert GW, Esler MD, Htun NM, Duval J, Hammond L, Eisenhardt SU, Flierl U, Schlaich MP, Peter K. Renal Denervation Reduces Monocyte Activation and Monocyte-Platelet Aggregate Formation: An Anti-Inflammatory Effect Relevant for Cardiovascular Risk. Hypertension 2016; 69:323-331. [PMID: 27956575 DOI: 10.1161/hypertensionaha.116.08373] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 09/17/2016] [Accepted: 11/16/2016] [Indexed: 02/06/2023]
Abstract
Overactivation of renal sympathetic nervous system and low-grade systemic inflammation are common features of hypertension. Renal denervation (RDN) reduces sympathetic activity in patients with resistant hypertension. However, its effect on systemic inflammation has not been examined. We prospectively investigated the effect of RDN on monocyte activation and inflammation in patients with uncontrolled hypertension scheduled for RDN. Ambulatory blood pressure, monocyte, and monocyte subset activation and inflammatory markers were assessed at baseline, 3 months, and 6 months after procedure in 42 patients. RDN significantly lowered blood pressure at 3 months (150.5±11.2/81.0±11.2 mm Hg to 144.7±11.8/77.9±11.0 mm Hg), which was sustained at 6 months (144.7±13.8/78.6±11.0 mm Hg). Activation status of monocytes significantly decreased at 3 months (P<0.01) and 6 months (P<0.01) after the procedure. In particular, classical monocyte activation was reduced at 6 months (P<0.05). Similarly, we observed a reduction of several inflammatory markers, including monocyte-platelet aggregates (3 months, P<0.01), plasma monocyte chemoattractant protein-1 levels (3 months, P<0.0001; 6 months, P<0.05), interleukin-1β (3 months, P<0.05; 6 months, P<0.05), tumor necrosis factor-α (3 months, P<0.01; 6 months, P<0.05), and interleukin-12 (3 months, P<0.01; 6 months, P<0.05). A positive correlation was observed between muscle sympathetic nerve activity and monocyte activation before and after the procedure. These results indicate that inhibition of sympathetic activity via RDN is associated with a reduction of monocyte activation and other inflammatory markers in hypertensive patients. These findings point to a direct interaction between the inflammatory and sympathetic nervous system, which is of central relevance for the understanding of beneficial cardiovascular effects of RDN.
Collapse
Affiliation(s)
- Maria T K Zaldivia
- From the Atherothrombosis and Vascular Biology (M.T.K.Z., J.R., B.L., N.M.H., S.U.E., U.F., K.P.) and Neurovascular Hypertension and Kidney Disease Laboratory (D.H., P.M., Y.S., N.E., R.L., G.W.L., M.D.E., J.D., L.H., M.P.S.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Melbourne (M.T.K.Z., N.M.H., M.P.S., K.P.); Dobney Hypertension Centre, School of Medicine and Pharmacology-Royal Perth Hospital Unit, University of Western Australia, Australia (D.H., P.M., M.P.S.); and Department of Plastic and Hand Surgery, University Medical Centre, Freiburg, Germany (S.U.E.)
| | - Jennifer Rivera
- From the Atherothrombosis and Vascular Biology (M.T.K.Z., J.R., B.L., N.M.H., S.U.E., U.F., K.P.) and Neurovascular Hypertension and Kidney Disease Laboratory (D.H., P.M., Y.S., N.E., R.L., G.W.L., M.D.E., J.D., L.H., M.P.S.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Melbourne (M.T.K.Z., N.M.H., M.P.S., K.P.); Dobney Hypertension Centre, School of Medicine and Pharmacology-Royal Perth Hospital Unit, University of Western Australia, Australia (D.H., P.M., M.P.S.); and Department of Plastic and Hand Surgery, University Medical Centre, Freiburg, Germany (S.U.E.)
| | - Dagmara Hering
- From the Atherothrombosis and Vascular Biology (M.T.K.Z., J.R., B.L., N.M.H., S.U.E., U.F., K.P.) and Neurovascular Hypertension and Kidney Disease Laboratory (D.H., P.M., Y.S., N.E., R.L., G.W.L., M.D.E., J.D., L.H., M.P.S.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Melbourne (M.T.K.Z., N.M.H., M.P.S., K.P.); Dobney Hypertension Centre, School of Medicine and Pharmacology-Royal Perth Hospital Unit, University of Western Australia, Australia (D.H., P.M., M.P.S.); and Department of Plastic and Hand Surgery, University Medical Centre, Freiburg, Germany (S.U.E.)
| | - Petra Marusic
- From the Atherothrombosis and Vascular Biology (M.T.K.Z., J.R., B.L., N.M.H., S.U.E., U.F., K.P.) and Neurovascular Hypertension and Kidney Disease Laboratory (D.H., P.M., Y.S., N.E., R.L., G.W.L., M.D.E., J.D., L.H., M.P.S.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Melbourne (M.T.K.Z., N.M.H., M.P.S., K.P.); Dobney Hypertension Centre, School of Medicine and Pharmacology-Royal Perth Hospital Unit, University of Western Australia, Australia (D.H., P.M., M.P.S.); and Department of Plastic and Hand Surgery, University Medical Centre, Freiburg, Germany (S.U.E.)
| | - Yusuke Sata
- From the Atherothrombosis and Vascular Biology (M.T.K.Z., J.R., B.L., N.M.H., S.U.E., U.F., K.P.) and Neurovascular Hypertension and Kidney Disease Laboratory (D.H., P.M., Y.S., N.E., R.L., G.W.L., M.D.E., J.D., L.H., M.P.S.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Melbourne (M.T.K.Z., N.M.H., M.P.S., K.P.); Dobney Hypertension Centre, School of Medicine and Pharmacology-Royal Perth Hospital Unit, University of Western Australia, Australia (D.H., P.M., M.P.S.); and Department of Plastic and Hand Surgery, University Medical Centre, Freiburg, Germany (S.U.E.)
| | - Bock Lim
- From the Atherothrombosis and Vascular Biology (M.T.K.Z., J.R., B.L., N.M.H., S.U.E., U.F., K.P.) and Neurovascular Hypertension and Kidney Disease Laboratory (D.H., P.M., Y.S., N.E., R.L., G.W.L., M.D.E., J.D., L.H., M.P.S.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Melbourne (M.T.K.Z., N.M.H., M.P.S., K.P.); Dobney Hypertension Centre, School of Medicine and Pharmacology-Royal Perth Hospital Unit, University of Western Australia, Australia (D.H., P.M., M.P.S.); and Department of Plastic and Hand Surgery, University Medical Centre, Freiburg, Germany (S.U.E.)
| | - Nina Eikelis
- From the Atherothrombosis and Vascular Biology (M.T.K.Z., J.R., B.L., N.M.H., S.U.E., U.F., K.P.) and Neurovascular Hypertension and Kidney Disease Laboratory (D.H., P.M., Y.S., N.E., R.L., G.W.L., M.D.E., J.D., L.H., M.P.S.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Melbourne (M.T.K.Z., N.M.H., M.P.S., K.P.); Dobney Hypertension Centre, School of Medicine and Pharmacology-Royal Perth Hospital Unit, University of Western Australia, Australia (D.H., P.M., M.P.S.); and Department of Plastic and Hand Surgery, University Medical Centre, Freiburg, Germany (S.U.E.)
| | - Rebecca Lee
- From the Atherothrombosis and Vascular Biology (M.T.K.Z., J.R., B.L., N.M.H., S.U.E., U.F., K.P.) and Neurovascular Hypertension and Kidney Disease Laboratory (D.H., P.M., Y.S., N.E., R.L., G.W.L., M.D.E., J.D., L.H., M.P.S.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Melbourne (M.T.K.Z., N.M.H., M.P.S., K.P.); Dobney Hypertension Centre, School of Medicine and Pharmacology-Royal Perth Hospital Unit, University of Western Australia, Australia (D.H., P.M., M.P.S.); and Department of Plastic and Hand Surgery, University Medical Centre, Freiburg, Germany (S.U.E.)
| | - Gavin W Lambert
- From the Atherothrombosis and Vascular Biology (M.T.K.Z., J.R., B.L., N.M.H., S.U.E., U.F., K.P.) and Neurovascular Hypertension and Kidney Disease Laboratory (D.H., P.M., Y.S., N.E., R.L., G.W.L., M.D.E., J.D., L.H., M.P.S.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Melbourne (M.T.K.Z., N.M.H., M.P.S., K.P.); Dobney Hypertension Centre, School of Medicine and Pharmacology-Royal Perth Hospital Unit, University of Western Australia, Australia (D.H., P.M., M.P.S.); and Department of Plastic and Hand Surgery, University Medical Centre, Freiburg, Germany (S.U.E.)
| | - Murray D Esler
- From the Atherothrombosis and Vascular Biology (M.T.K.Z., J.R., B.L., N.M.H., S.U.E., U.F., K.P.) and Neurovascular Hypertension and Kidney Disease Laboratory (D.H., P.M., Y.S., N.E., R.L., G.W.L., M.D.E., J.D., L.H., M.P.S.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Melbourne (M.T.K.Z., N.M.H., M.P.S., K.P.); Dobney Hypertension Centre, School of Medicine and Pharmacology-Royal Perth Hospital Unit, University of Western Australia, Australia (D.H., P.M., M.P.S.); and Department of Plastic and Hand Surgery, University Medical Centre, Freiburg, Germany (S.U.E.)
| | - Nay M Htun
- From the Atherothrombosis and Vascular Biology (M.T.K.Z., J.R., B.L., N.M.H., S.U.E., U.F., K.P.) and Neurovascular Hypertension and Kidney Disease Laboratory (D.H., P.M., Y.S., N.E., R.L., G.W.L., M.D.E., J.D., L.H., M.P.S.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Melbourne (M.T.K.Z., N.M.H., M.P.S., K.P.); Dobney Hypertension Centre, School of Medicine and Pharmacology-Royal Perth Hospital Unit, University of Western Australia, Australia (D.H., P.M., M.P.S.); and Department of Plastic and Hand Surgery, University Medical Centre, Freiburg, Germany (S.U.E.)
| | - Jacqueline Duval
- From the Atherothrombosis and Vascular Biology (M.T.K.Z., J.R., B.L., N.M.H., S.U.E., U.F., K.P.) and Neurovascular Hypertension and Kidney Disease Laboratory (D.H., P.M., Y.S., N.E., R.L., G.W.L., M.D.E., J.D., L.H., M.P.S.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Melbourne (M.T.K.Z., N.M.H., M.P.S., K.P.); Dobney Hypertension Centre, School of Medicine and Pharmacology-Royal Perth Hospital Unit, University of Western Australia, Australia (D.H., P.M., M.P.S.); and Department of Plastic and Hand Surgery, University Medical Centre, Freiburg, Germany (S.U.E.)
| | - Louise Hammond
- From the Atherothrombosis and Vascular Biology (M.T.K.Z., J.R., B.L., N.M.H., S.U.E., U.F., K.P.) and Neurovascular Hypertension and Kidney Disease Laboratory (D.H., P.M., Y.S., N.E., R.L., G.W.L., M.D.E., J.D., L.H., M.P.S.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Melbourne (M.T.K.Z., N.M.H., M.P.S., K.P.); Dobney Hypertension Centre, School of Medicine and Pharmacology-Royal Perth Hospital Unit, University of Western Australia, Australia (D.H., P.M., M.P.S.); and Department of Plastic and Hand Surgery, University Medical Centre, Freiburg, Germany (S.U.E.)
| | - Steffen U Eisenhardt
- From the Atherothrombosis and Vascular Biology (M.T.K.Z., J.R., B.L., N.M.H., S.U.E., U.F., K.P.) and Neurovascular Hypertension and Kidney Disease Laboratory (D.H., P.M., Y.S., N.E., R.L., G.W.L., M.D.E., J.D., L.H., M.P.S.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Melbourne (M.T.K.Z., N.M.H., M.P.S., K.P.); Dobney Hypertension Centre, School of Medicine and Pharmacology-Royal Perth Hospital Unit, University of Western Australia, Australia (D.H., P.M., M.P.S.); and Department of Plastic and Hand Surgery, University Medical Centre, Freiburg, Germany (S.U.E.)
| | - Ulrike Flierl
- From the Atherothrombosis and Vascular Biology (M.T.K.Z., J.R., B.L., N.M.H., S.U.E., U.F., K.P.) and Neurovascular Hypertension and Kidney Disease Laboratory (D.H., P.M., Y.S., N.E., R.L., G.W.L., M.D.E., J.D., L.H., M.P.S.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Melbourne (M.T.K.Z., N.M.H., M.P.S., K.P.); Dobney Hypertension Centre, School of Medicine and Pharmacology-Royal Perth Hospital Unit, University of Western Australia, Australia (D.H., P.M., M.P.S.); and Department of Plastic and Hand Surgery, University Medical Centre, Freiburg, Germany (S.U.E.)
| | - Markus P Schlaich
- From the Atherothrombosis and Vascular Biology (M.T.K.Z., J.R., B.L., N.M.H., S.U.E., U.F., K.P.) and Neurovascular Hypertension and Kidney Disease Laboratory (D.H., P.M., Y.S., N.E., R.L., G.W.L., M.D.E., J.D., L.H., M.P.S.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Melbourne (M.T.K.Z., N.M.H., M.P.S., K.P.); Dobney Hypertension Centre, School of Medicine and Pharmacology-Royal Perth Hospital Unit, University of Western Australia, Australia (D.H., P.M., M.P.S.); and Department of Plastic and Hand Surgery, University Medical Centre, Freiburg, Germany (S.U.E.)
| | - Karlheinz Peter
- From the Atherothrombosis and Vascular Biology (M.T.K.Z., J.R., B.L., N.M.H., S.U.E., U.F., K.P.) and Neurovascular Hypertension and Kidney Disease Laboratory (D.H., P.M., Y.S., N.E., R.L., G.W.L., M.D.E., J.D., L.H., M.P.S.), Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Melbourne (M.T.K.Z., N.M.H., M.P.S., K.P.); Dobney Hypertension Centre, School of Medicine and Pharmacology-Royal Perth Hospital Unit, University of Western Australia, Australia (D.H., P.M., M.P.S.); and Department of Plastic and Hand Surgery, University Medical Centre, Freiburg, Germany (S.U.E.).
| |
Collapse
|
30
|
Cottin SC, Alsaleh A, Sanders TAB, Hall WL. Lack of effect of supplementation with EPA or DHA on platelet-monocyte aggregates and vascular function in healthy men. Nutr Metab Cardiovasc Dis 2016; 26:743-751. [PMID: 27105870 DOI: 10.1016/j.numecd.2016.03.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 03/07/2016] [Accepted: 03/08/2016] [Indexed: 11/21/2022]
Abstract
BACKGROUND AND AIMS Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) found in fish oil are postulated to have favourable effects on platelet, endothelial and vascular function. We investigated whether EPA has differential effects on in vivo platelet aggregation and other markers of cardiovascular risk compared to DHA. METHODS AND RESULTS Following a 2 wk run-in taking encapsulated refined olive oil, 48 healthy young men were randomly allocated using a parallel design to receive EPA-rich (3.1 g EPA/d) or DHA-rich (2.9 g DHA/d) triglyceride concentrates or refined olive oil (placebo), for a total supplementary lipid intake of 5 g/d. The specified primary outcome was change in platelet monocyte aggregates (PMA); secondary outcomes were capillary density, augmentation index, digital pulse volume measurements, 24 h ambulatory BP, plasma 8-isoprostanes-F2α. Changes in the proportions of DHA and EPA in erythrocytes and non-esterified fatty acid composition indicated compliance to the intervention. There was no significant treatment effect on PMA (P = 0.382); mean changes (%) (95% CI) were placebo -0.5 (-2.0, 1.04), EPA 0.4 (-0.8, 1.6), DHA 0.3 (-1.5, 2.0). R-QUICKI, an index of insulin sensitivity, was greater following EPA compared to placebo (P < 0.05). No other significant differences were noted. CONCLUSION Neither EPA- nor DHA-rich fish oil supplementation influence platelet-monocyte aggregation or several markers of vascular function after 6 wk in healthy young males. This trial was registered at clinicaltrials.gov as NCT01735357.
Collapse
Affiliation(s)
- S C Cottin
- From King's College London, Diabetes and Nutritional Sciences Division, Faculty of Life Sciences & Medicine, 150 Stamford Street, London, SE1 9NH, UK.
| | - A Alsaleh
- From King's College London, Diabetes and Nutritional Sciences Division, Faculty of Life Sciences & Medicine, 150 Stamford Street, London, SE1 9NH, UK
| | - T A B Sanders
- From King's College London, Diabetes and Nutritional Sciences Division, Faculty of Life Sciences & Medicine, 150 Stamford Street, London, SE1 9NH, UK
| | - W L Hall
- From King's College London, Diabetes and Nutritional Sciences Division, Faculty of Life Sciences & Medicine, 150 Stamford Street, London, SE1 9NH, UK
| |
Collapse
|
31
|
Gkaliagkousi E, Gavriilaki E, Triantafyllou A, Douma S. Clinical Significance of Endothelial Dysfunction in Essential Hypertension. Curr Hypertens Rep 2016; 17:85. [PMID: 26371063 DOI: 10.1007/s11906-015-0596-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The endothelium is recognized as a major determinant of vascular physiology and pathophysiology. Over the last few decades, a plethora of studies have implicated endothelial dysfunction in the progression of atherosclerosis and the subclinical target organ damage observed in essential hypertension. However, the clinical significance of diagnosing endothelial dysfunction in patients with essential hypertension remains under investigation. Although a number of vascular and non-vascular markers of endothelial dysfunction have been proposed, there is an ongoing quest for a marker in the clinical setting that is optimal, inexpensive, and reproducible. In addition, endothelial dysfunction emerges as a promising therapeutic target of agents that are readily available in clinical practice. In this context, a better understanding of its role in essential hypertension becomes of great importance. Here, we aim to investigate the clinical significance of endothelial dysfunction in essential hypertension by accumulating novel data on (a) early diagnosis using robust markers with prognostic value in cardiovascular risk prediction, (b) the association of endothelial dysfunction with subclinical vascular organ damage, and (c) potential therapeutic targets.
Collapse
Affiliation(s)
- Eugenia Gkaliagkousi
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Aristotle University of Thessaloniki, Ring Road Nea Eukarpia, 564 03, Thessaloniki, Greece.
| | - Eleni Gavriilaki
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Aristotle University of Thessaloniki, Ring Road Nea Eukarpia, 564 03, Thessaloniki, Greece
| | - Areti Triantafyllou
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Aristotle University of Thessaloniki, Ring Road Nea Eukarpia, 564 03, Thessaloniki, Greece
| | - Stella Douma
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Aristotle University of Thessaloniki, Ring Road Nea Eukarpia, 564 03, Thessaloniki, Greece
| |
Collapse
|
32
|
Glezeva N, Gilmer JF, Watson CJ, Ledwidge M. A Central Role for Monocyte-Platelet Interactions in Heart Failure. J Cardiovasc Pharmacol Ther 2015; 21:245-61. [PMID: 26519384 DOI: 10.1177/1074248415609436] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 08/04/2015] [Indexed: 01/08/2023]
Abstract
Heart failure (HF) is an increasingly prevalent and costly multifactorial syndrome with high morbidity and mortality rates. The exact pathophysiological mechanisms leading to the development of HF are not completely understood. Several emerging paradigms implicate cardiometabolic risk factors, inflammation, endothelial dysfunction, myocardial fibrosis, and myocyte dysfunction as key factors in the gradual progression from a healthy state to HF. Inflammation is now a recognized factor in disease progression in HF and a therapeutic target. Furthermore, the monocyte-platelet interaction has been highlighted as an important pathophysiological link between inflammation, thrombosis, endothelial activation, and myocardial malfunction. The contribution of monocytes and platelets to acute cardiovascular injury and acute HF is well established. However, their role and interaction in the pathogenesis of chronic HF are not well understood. In particular, the cross talk between monocytes and platelets in the peripheral circulation and in the vicinity of the vascular wall in the form of monocyte-platelet complexes (MPCs) may be a crucial element, which influences the pathophysiology and progression of chronic heart disease and HF. In this review, we discuss the role of monocytes and platelets as key mediators of cardiovascular inflammation in HF, the mechanisms of cell activation, and the importance of monocyte-platelet interaction and complexes in HF pathogenesis. Finally, we summarize recent information on pharmacological inhibition of inflammation and studies of antithrombotic strategies in the setting of HF that can inform opportunities for future work. We discuss recent data on monocyte-platelet interactions and the potential benefits of therapy directed at MPCs, particularly in the setting of HF with preserved ejection fraction.
Collapse
Affiliation(s)
- Nadezhda Glezeva
- School of Medicine & Medical Science, UCD Conway Institute, University College Dublin, Dublin, Belfield, Dublin, Ireland
| | - John F Gilmer
- School of Pharmacy & Pharmaceutical Sciences, TCD Centre for Health Sciences, Trinity College Dublin, College Green, Dublin, Ireland
| | - Chris J Watson
- School of Medicine & Medical Science, UCD Conway Institute, University College Dublin, Dublin, Belfield, Dublin, Ireland
| | - Mark Ledwidge
- Chronic Cardiovascular Disease Management Unit and Heart Failure Unit, St Vincent's Healthcare Group/St Michael's Hospital, County Dublin, Ireland
| |
Collapse
|
33
|
Ed Rainger G, Chimen M, Harrison MJ, Yates CM, Harrison P, Watson SP, Lordkipanidzé M, Nash GB. The role of platelets in the recruitment of leukocytes during vascular disease. Platelets 2015. [PMID: 26196409 PMCID: PMC4673595 DOI: 10.3109/09537104.2015.1064881] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Besides their role in the formation of thrombus during haemostasis, it is becoming clear that platelets contribute to a number of other processes within the vasculature. Indeed, the integrated function of the thrombotic and inflammatory systems, which results in platelet-mediated recruitment of leukocytes, is now considered to be of great importance in the propagation, progression and pathogenesis of atherosclerotic disease of the arteries. There are three scenarios by which platelets can interact with leukocytes: (1) during haemostasis, when platelets adhere to and are activated on sub-endothelial matrix proteins exposed by vascular damage and then recruit leukocytes to a growing thrombus. (2) Platelets adhere to and are activated on stimulated endothelial cells and then bridge blood borne leukocytes to the vessel wall and. (3) Adhesion between platelets and leukocytes occurs in the blood leading to formation of heterotypic aggregates prior to contact with endothelial cells. In the following review we will not discuss leukocyte recruitment during haemostasis, as this represents a physiological response to tissue trauma that can progress, at least in its early stages, in the absence of inflammation. Rather we will deal with scenarios 2 and 3, as these pathways of platelet–leukocyte interactions are important during inflammation and in chronic inflammatory diseases such as atherosclerosis. Indeed, these interactions mean that leukocytes possess means of adhesion to the vessel wall under conditions that may not normally be permissive of leukocyte–endothelial cell adhesion, meaning that the disease process may be able to bypass the regulatory pathways which would ordinarily moderate the inflammatory response.
Collapse
Affiliation(s)
- G Ed Rainger
- Centre for Cardiovascular Sciences, Institute for Biomedical Research, The Medical School, The University of Birmingham , Birmingham , UK and
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Gambaryan S, Tsikas D. A review and discussion of platelet nitric oxide and nitric oxide synthase: do blood platelets produce nitric oxide from L-arginine or nitrite? Amino Acids 2015; 47:1779-93. [PMID: 25929585 DOI: 10.1007/s00726-015-1986-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 04/09/2015] [Indexed: 02/07/2023]
Abstract
The NO/sGC/cGMP/PKG system is one of the most powerful mechanisms responsible for platelet inhibition. In numerous publications, expression of functional NO synthase (NOS) in human and mouse platelets has been reported. Constitutive and inducible NOS isoforms convert L-arginine to NO and L-citrulline. The importance of this pathway in platelets and in endothelial cells for the regulation of platelet function is discussed since decades. However, there are serious doubts in the literature concerning both expression and functionality of NOS in platelets. In this review, we aim to present and critically evaluate recent data concerning NOS expression and function in platelets, and to especially emphasise potential pitfalls of detection of NOS proteins and measurement of NOS activity. Prevailing analytical problems are probably the main sources of contradictory data on occurrence, activity and function of NOS in platelets. In this review we also address issues of how these problems can be resolved. NO donors including organic nitrites (RONO) and organic nitrate (RONO2) are inhibitors of platelet activation. Endogenous inorganic nitrite (NO2 (-)), the product of NO autoxidation, and exogenous inorganic nitrite are increasingly investigated as NO donors in the circulation. The role of platelets in the generation of NO from nitrite is also discussed.
Collapse
Affiliation(s)
- Stepan Gambaryan
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez Prosp, St. Petersburg, 194223, Russia,
| | | |
Collapse
|
35
|
Murray KN, Parry-Jones AR, Allan SM. Interleukin-1 and acute brain injury. Front Cell Neurosci 2015; 9:18. [PMID: 25705177 PMCID: PMC4319479 DOI: 10.3389/fncel.2015.00018] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 01/12/2015] [Indexed: 01/05/2023] Open
Abstract
Inflammation is the key host-defense response to infection and injury, yet also a major contributor to a diverse range of diseases, both peripheral and central in origin. Brain injury as a result of stroke or trauma is a leading cause of death and disability worldwide, yet there are no effective treatments, resulting in enormous social and economic costs. Increasing evidence, both preclinical and clinical, highlights inflammation as an important factor in stroke, both in determining outcome and as a contributor to risk. A number of inflammatory mediators have been proposed as key targets for intervention to reduce the burden of stroke, several reaching clinical trial, but as yet yielding no success. Many factors could explain these failures, including the lack of robust preclinical evidence and poorly designed clinical trials, in addition to the complex nature of the clinical condition. Lack of consideration in preclinical studies of associated co-morbidities prevalent in the clinical stroke population is now seen as an important omission in previous work. These co-morbidities (atherosclerosis, hypertension, diabetes, infection) have a strong inflammatory component, supporting the need for greater understanding of how inflammation contributes to acute brain injury. Interleukin (IL)-1 is the prototypical pro-inflammatory cytokine, first identified many years ago as the endogenous pyrogen. Research over the last 20 years or so reveals that IL-1 is an important mediator of neuronal injury and blocking the actions of IL-1 is beneficial in a number of experimental models of brain damage. Mechanisms underlying the actions of IL-1 in brain injury remain unclear, though increasing evidence indicates the cerebrovasculature as a key target. Recent literature supporting this and other aspects of how IL-1 and systemic inflammation in general contribute to acute brain injury are discussed in this review.
Collapse
Affiliation(s)
- Katie N Murray
- Faculty of Life Sciences, University of Manchester Manchester, UK
| | | | - Stuart M Allan
- Faculty of Life Sciences, University of Manchester Manchester, UK
| |
Collapse
|
36
|
Gkaliagkousi E, Gavriilaki E, Yiannaki E, Markala D, Papadopoulos N, Triantafyllou A, Anyfanti P, Petidis K, Garypidou V, Doumas M, Ferro A, Douma S. Platelet activation in essential hypertension during exercise: pre- and post-treatment changes with an angiotensin II receptor blocker. Am J Hypertens 2014; 27:571-8. [PMID: 23975222 DOI: 10.1093/ajh/hpt153] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Acute exercise may exert deleterious effects on the cardiovascular system through a variety of pathophysiological mechanisms, including increased platelet activation. However, the degree of exercise-induced platelet activation in untreated hypertensive (UH) individuals as compared with normotensive (NT) individuals has yet to be established. Furthermore, the effect of antihypertensive treatment on exercise-induced platelet activation in essential hypertension (EH) remains unknown. METHODS Study 1 consisted of 30 UH and 15 NT subjects. UH subjects who received treatment were included in study 2 and were followed-up after a 3-month treatment period with an angiotensin II receptor blocker (ARB; valsartan). Circulating monocyte-platelet aggregates (MPA) and platelet P-selectin were measured as platelet activation markers at baseline, immediately after a treadmill exercise test, and 10, 30, and 90 minutes later. RESULTS Maximal platelet activation was observed at 10 minutes after peak exercise in both groups. In UH subjects, MPA levels remained increased at 30 minutes after peak exercise, despite BP fall to baseline levels. MPA levels were significantly higher in UH subjects than NT subjects at maximal exercise and at 10 and 30 minutes of recovery. Post-treatment MPA levels increased significantly only at 10 minutes into recovery and were similar to those of NT subjects. CONCLUSIONS Acute high-intensity exercise exaggerates platelet activation in untreated patients with EH compared with NT individuals. Angiotensin II receptor blockade with adequate BP control greatly improves exercise-induced platelet activation in EH. Further studies are needed to clarify whether this phenomenon depends purely on BP lowering or benefits also from the pleiotropic effects of ARBs.
Collapse
Affiliation(s)
- Eugenia Gkaliagkousi
- 2nd Propedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Czepluch FS, Kuschicke H, Dellas C, Riggert J, Hasenfuss G, Schäfer K. Increased proatherogenic monocyte-platelet cross-talk in monocyte subpopulations of patients with stable coronary artery disease. J Intern Med 2014; 275:144-54. [PMID: 24118494 DOI: 10.1111/joim.12145] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Monocytes and platelets are important cellular mediators of atherosclerosis. Human monocytes can be divided into CD14(++) CD16(-) , CD14(++) CD16(+) and CD14(+) CD16(++) cells, which differ in their functional properties. The aim of this study was to examine monocyte subset distribution, monocyte-platelet aggregate (MPA) formation and expression of CCR5, the receptor of the platelet-derived chemokine CCL5, and to determine whether these parameters are altered in individuals with coronary atherosclerosis. METHODS Peripheral blood cells from 64 healthy blood donors (HBDs) and 60 patients with stable coronary artery disease (CAD) were stained with antibodies against CD14, CD16, CD42b and CCR5 and analysed by flow cytometry. Circulating CCL5 levels were determined using an enzyme-linked immunosorbent assay. RESULTS In patients with CAD, the relative proportion of the CD14(++) CD16(-) monocyte subset was elevated (P < 0.05) and of the CD14(+) CD16(++) subset was reduced (P < 0.001) compared with the HBD group. Furthermore, MPA formation significantly increased in patients with CAD in all three monocyte subsets. In both study groups, the majority of CCR5(+) cells was detected in CD14(++) CD16(+) monocytes (P < 0.001 versus CD14(++) CD16(-) and CD14(+) CD16(++) ), although the CCR5(+) monocyte number was reduced in patients with CAD (CD14(++) CD16(-) /CD14(+) CD16(++) , P < 0.001; CD14(++) CD16(+) , P < 0.05) compared with the HBD group, particularly in those who were not taking statins. Ex vivo incubation of monocytes from HBDs with plasma from patients with CAD also decreased CCR5(+) expression (P < 0.05 versus plasma from HBDs). Serum CCL5 levels were similar in both groups. CONCLUSIONS The increased monocyte-platelet cross-talk in patients with CAD might have contributed to atherosclerosis progression. The decreased CCR5(+) monocyte numbers in patients with CAD could have resulted from CCR5(+) cell recruitment into atherosclerotic lesions or CCR5 downregulation in response to circulating factors.
Collapse
Affiliation(s)
- F S Czepluch
- Department of Cardiology and Pulmonary Medicine, University Medical Center Göttingen, Göttingen, Germany
| | | | | | | | | | | |
Collapse
|
38
|
Gavriilaki E, Gkaliagkousi E, Nikolaidou B, Douma S. Mean platelet volume in hypertensive urgencies and emergencies: opening the Pandora's Box. Platelets 2014; 26:275-6. [PMID: 24433202 DOI: 10.3109/09537104.2013.868426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Eleni Gavriilaki
- 2nd Propedeutic Department of Internal Medicine, Aristotle University of Thessaloniki , Thessaloniki , Greece and
| | | | | | | |
Collapse
|
39
|
Exercise-induced platelet activation in essential hypertension: have we solved the puzzle? J Hypertens 2013; 31:1915-6. [PMID: 24107673 DOI: 10.1097/hjh.0b013e328363e811] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
40
|
Kolarova H, Klinke A, Kremserova S, Adam M, Pekarova M, Baldus S, Eiserich JP, Kubala L. Myeloperoxidase induces the priming of platelets. Free Radic Biol Med 2013; 61:357-69. [PMID: 23603662 DOI: 10.1016/j.freeradbiomed.2013.04.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2012] [Revised: 04/10/2013] [Accepted: 04/11/2013] [Indexed: 12/23/2022]
Abstract
The release of myeloperoxidase (MPO) from polymorphonuclear neutrophils is a hallmark of vascular inflammation and contributes to the pathogenesis of vascular inflammatory processes. However, the effects of MPO on platelets as a contributory mechanism in vascular inflammatory diseases remain unknown. Thus, MPO interaction with platelets and its effects on platelet function were examined. First, dose-dependent binding of MPO (between 1.7 and 13.8nM) to both human and mouse platelets was observed. This was in direct contrast to the absence of MPO in megakaryocytes. MPO was localized both on the surface of and inside platelets. Cytoskeleton inhibition did not prevent MPO localization inside the three-dimensional platelet structure. MPO peroxidase activity was preserved upon the MPO binding to platelets. MPO sequestered in platelets catabolized NO, documented by the decreased production of NO (on average, an approximately 2-fold decrease). MPO treatment did not affect the viability of platelets during short incubations; however, it decreased platelet viability after long-term storage for 7 days (an approximately 2-fold decrease). The activation of platelets by MPO was documented by an MPO-mediated increase in the expression of surface platelet receptors P-selectin and PECAM-1 (of about 5 to 20%) and the increased formation of reactive oxygen species (of about 15 to 200%). However, the activation was only partial, as MPO did not induce the aggregation of platelets nor potentiate platelet response to classical activators. Nor did MPO induce a significant release of the content of granules. The activation of platelets by MPO was connected with increased MPO-treated platelet interaction with polymorphonuclear leukocytes (an approximately 1.2-fold increase) in vitro. In conclusion, it can be suggested that MPO can interact with and activate platelets, which can induce priming of platelets, rather than the classical robust activation of platelets. This can contribute to the development of chronic inflammatory processes in vessels.
Collapse
Affiliation(s)
- H Kolarova
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czech Republic; Department of Animal Physiology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - A Klinke
- Department of Cardiology, University Heart Center Hamburg, University Hospital Eppendorf, Hamburg, Germany
| | - S Kremserova
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czech Republic; Department of Animal Physiology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - M Adam
- Department of Cardiology, University Heart Center Hamburg, University Hospital Eppendorf, Hamburg, Germany
| | - M Pekarova
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czech Republic
| | - S Baldus
- Department of Cardiology, University Heart Center Hamburg, University Hospital Eppendorf, Hamburg, Germany
| | - J P Eiserich
- Division of Pulmonary/Critical Care Medicine, Department of Internal Medicine, School of Medicine, University of California at Davis, Davis, CA, USA
| | - L Kubala
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czech Republic; International Clinical Research Center-Center of Biomolecular and Cellular Engineering, St. Anne's University Hospital Brno, Brno, Czech Republic.
| |
Collapse
|
41
|
Gianfagna F, Tamburrelli C, Vohnout B, Crescente M, Izzi B, Pampuch A, De Curtis A, Di Castelnuovo A, Cutrone A, Napoleone E, Tayo B, Lorenzet R, Nanni L, Arca M, Donati MB, de Gaetano G, Cerletti C, Iacoviello L. Heritability, genetic correlation and linkage to the 9p21.3 region of mixed platelet-leukocyte conjugates in families with and without early myocardial infarction. Nutr Metab Cardiovasc Dis 2013; 23:684-692. [PMID: 22633792 DOI: 10.1016/j.numecd.2012.02.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Revised: 02/02/2012] [Accepted: 02/27/2012] [Indexed: 11/25/2022]
Abstract
BACKGROUND AND AIMS Variations in mixed platelet-leukocyte conjugate formation in human whole blood could be genetically determined. We quantified platelet and leukocyte activation and interaction in families with or without early myocardial infarction and evaluated their heritability, genetic correlation and linkage to the 9p21.3 region. METHODS AND RESULTS The study population included 739 subjects (≥ 15 years old) from 54 large pedigrees, 23 with and 31 without familial myocardial infarction. Mixed platelet-leukocyte conjugates and markers of platelet or leukocyte activation (P-selectin, CD11b and L-selectin surface expression) were measured both before and after in vitro blood stimulation with collagen-ADP. All traits had significant genetic components (17.5-65.3% of the phenotypic variability), while shared household effects (0-39.6%) and environmental covariates (0-10.2%) tended to be smaller. Stimulated platelet-polymorphonuclear leukocyte (PMN) and platelet-monocyte conjugates showed the highest linkage to the 9p21.3 region (LOD = 0.94 and 1.33, respectively; empirical p value = 0.017 and 0.009). PMN markers resulted strongly genetically correlated between them in bivariate analysis among pairs of quantitative traits. CONCLUSION This study supports a genetic regulation of human mixed platelet-leukocyte conjugates.
Collapse
Affiliation(s)
- F Gianfagna
- Laboratory of Genetic and Environmental Epidemiology, Fondazione di Ricerca e Cura Giovanni Paolo II, Università Cattolica, Campobasso, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Murray KN, Buggey HF, Denes A, Allan SM. Systemic immune activation shapes stroke outcome. Mol Cell Neurosci 2012; 53:14-25. [PMID: 23026562 DOI: 10.1016/j.mcn.2012.09.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 09/11/2012] [Accepted: 09/21/2012] [Indexed: 02/07/2023] Open
Abstract
Stroke is a major cause of morbidity and mortality, and activation of the immune system can impact on stroke outcome. Although the majority of research has focused on the role of the immune system after stroke there is increasing evidence to suggest that inflammation and immune activation prior to brain injury can influence stroke risk and outcome. With the high prevalence of co-morbidities in the Western world such as obesity, hypertension and diabetes, pre-existing chronic 'low-grade' systemic inflammation has become a customary characteristic of stroke pathophysiology that needs to be considered in the search for new therapies. The importance of the immune system in stroke has been demonstrated in a number of ways, both experimentally and in the clinical setting. This review will focus on the effect of immune activation arising from systemic inflammatory conditions and infection, how it affects the incidence and outcomes of stroke, and the possible underlying mechanisms involved. This article is part of a Special Issue entitled 'Neuroinflammation in neurodegeneration and neurodysfunction'.
Collapse
Affiliation(s)
- Katie N Murray
- Faculty of Life Sciences, A.V. Hill Building, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | | | | | | |
Collapse
|
43
|
Yu JG, Zhou RR, Cai GJ. From hypertension to stroke: mechanisms and potential prevention strategies. CNS Neurosci Ther 2012; 17:577-84. [PMID: 21951373 DOI: 10.1111/j.1755-5949.2011.00264.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Stroke is a major cause of disability and death worldwide. Prevention aimed at risk factors of stroke is the most effective strategy to curb the stroke pandemic. Hypertension is one of the most important risk factors for stroke. Despite the substantial evidence of the benefits of lowering blood pressure, conventional treatment does not normalize the burden of major cardiovascular events in patients with hypertension. Fully understanding the factors involved in the hypertension-induced stroke helps to develop new strategies for stroke prevention. Antihypertensive therapies selected should have positive blood pressure-independent effects on stroke risk. This review summarizes the factors involved in the hypertension-induced stroke, such as oxidative stress, inflammation, and arterial baroreflex dysfunction, and potential strategies for its prevention, therefore, provides clues for clinicians.
Collapse
Affiliation(s)
- Jian-Guang Yu
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | | | | |
Collapse
|
44
|
Detection of circulating platelet-monocyte complexes in persons infected with human immunodeficiency virus type-1. J Virol Methods 2012; 181:170-6. [PMID: 22387340 DOI: 10.1016/j.jviromet.2012.02.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Revised: 02/03/2012] [Accepted: 02/13/2012] [Indexed: 11/24/2022]
Abstract
Activated platelets form transient aggregates with monocytes in circulation and have a half-life of approximately 30-60 min. These complexes are increased in various inflammatory conditions and are an early marker of myocardial infarction. HIV-1 infection is associated with chronic inflammation, and increased CD16⁺ inflammatory monocytes have been observed in these individuals, probably as a result of increased interaction with platelets. However, narrow detection period and platelet activation during sample processing pose significant problems in detecting platelet-monocyte complexes (PMCs). A method was standardized addressing these difficulties, to enumerate PMCs involving CD16⁺ or CD16⁻ monocytes in whole blood using flow cytometry. Blood collected from healthy individuals was treated with either collagen (for platelet activation) or LPS (for monocyte activation) and subsequently used to study effect of these treatments on PMC formation. This method was also validated for the ex vivo quantitation of PMCs in blood obtained from persons infected with HIV. The in vitro results demonstrated that platelet activation, but not monocyte activation, resulted in significant increase in PMC formation. There was a significant increase in CD16⁺ PMCs and platelet activation, in samples obtained from persons infected with HIV as compared to those without HIV infection. Furthermore, PMC percentages correlated positively with platelet activation. These findings improve the ability to detect PMCs and shed light on HIV pathogenesis.
Collapse
|
45
|
Passacquale G, Ferro A. Current concepts of platelet activation: possibilities for therapeutic modulation of heterotypic vs. homotypic aggregation. Br J Clin Pharmacol 2012; 72:604-18. [PMID: 21223359 DOI: 10.1111/j.1365-2125.2011.03906.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Thrombogenic and inflammatory activity are two distinct aspects of platelet biology, which are sustained by the ability of activated platelets to interact with each other (homotypic aggregation) and to adhere to circulating leucocytes (heterotypic aggregation). These two events are regulated by distinct biomolecular mechanisms that are selectively activated in different pathophysiological settings. They can occur simultaneously, for example, as part of a pro-thrombotic/pro-inflammatory response induced by vascular damage, or independently, as in certain clinical conditions in which abnormal heterotypic aggregation has been observed in the absence of intravascular thrombosis. Current antiplatelet drugs have been developed to target specific molecular signalling pathways mainly implicated in thrombus formation, and their ever increasing clinical use has resulted in clear benefits in the treatment and prevention of arterial thrombotic events. However, the efficacy of currently available antiplatelet drugs remains suboptimal, most likely because their therapeutic action is limited to only few of the signalling pathways involved in platelet homotypic aggregation. In this context, modulation of heterotypic aggregation, which is believed to contribute importantly to acute thrombotic events, as well to the pathophysiology of atherosclerosis itself, may offer benefits over and above the classical antiplatelet approach. This review will focus on the distinct biomolecular pathways that, following platelet activation, underlie homotypic and heterotypic aggregation, aiming potentially to identify novel therapeutic targets.
Collapse
Affiliation(s)
- Gabriella Passacquale
- Department of Clinical Pharmacology, Cardiovascular Division, King's College London, London, UK
| | | |
Collapse
|
46
|
Alexandru N, Popov D, Georgescu A. Platelet dysfunction in vascular pathologies and how can it be treated. Thromb Res 2011; 129:116-26. [PMID: 22035630 DOI: 10.1016/j.thromres.2011.09.026] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Revised: 09/19/2011] [Accepted: 09/22/2011] [Indexed: 12/17/2022]
Abstract
Cardiovascular diseases are one of the leading causes of morbidity and mortality in industrialized countries, and although many processes play a role in the development of vascular disease, thrombosis is the primary event that precipitates stroke and acute coronary syndromes. The blood platelets are of significant importance in medicine. These cells are involved in many physiological processes, particularly haemostasis through their ability to aggregate and form clots in response to activation. In addition, these dynamic cells display activities that extend beyond thrombosis, including an important role in initiating and sustaining vascular inflammation. The expansion of knowledge from basic and clinical research has highlighted the critical position of platelets in several inflammatory diseases such as arthritis and atherosclerosis. Platelets are emerging as important mediators of inflammation and provide important signals to mediate phenotype of other blood and vascular cells. The important role of platelets in arterial thrombosis and the onset of acute myocardial infarction after atherosclerotic plaque rupture make inhibition of platelet aggregation a critical step in preventing thrombotic events associated with stroke, heart attack, and peripheral arterial thrombosis. However, the use of platelet inhibitors for thrombosis prevention must seek a delicate balance between inhibiting platelet activation and an associated increased bleeding risk. The aim of this review is to up-date the knowledge on platelets physiology and dysfunction in pathologies, such as diabetes mellitus, hypercholesterolemia, and hypertension, emphasizing the link between platelets and the inflammation-related atherosclerosis. The review evaluates the opportunities offered by the novel platelet inhibitors to efficiently alleviate the thrombotic events.
Collapse
|
47
|
Passacquale G, Vamadevan P, Pereira L, Hamid C, Corrigall V, Ferro A. Monocyte-platelet interaction induces a pro-inflammatory phenotype in circulating monocytes. PLoS One 2011; 6:e25595. [PMID: 22022418 PMCID: PMC3192052 DOI: 10.1371/journal.pone.0025595] [Citation(s) in RCA: 145] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 09/06/2011] [Indexed: 01/07/2023] Open
Abstract
Background Activated platelets exert a pro-inflammatory action that can be largely ascribed to their ability to interact with leukocytes and modulate their activity. We hypothesized that platelet activation and consequent formation of monocyte-platelet aggregates (MPA) induces a pro-inflammatory phenotype in circulating monocytes. Methodology/Principal Findings CD62P+ platelets and MPA were measured, and monocytes characterized, by whole blood flow cytometry in healthy subjects, before and two days after receiving influenza immunization. Three monocytic subsets were identified: CD14+CD16−, CD14highCD16+and CD14lowCD16+. The increase in high sensitivity C-reactive protein post-immunization was accompanied by increased platelet activation and MPA formation (25.02±12.57 vs 41.48±16.81; p = 0.01), along with enhancement of circulating CD14highCD16+ cells (4.7±3.6 vs 10.4±4.8; p = 0.003), their percentage being linearly related to levels of CD62P+-platelets (r2 = 0.4347; p = 0.0008). In separate in vitro experiments, co-incubation of CD14+CD16− cells, isolated from healthy donor subjects, with autologous platelets gave rise to up-regulation of CD16 on monocytes as compared with those maintained in medium alone (% change in CD14+CD16+ cells following 48 h co-incubation of monocytes with platelets was +106±51% vs monocytes in medium alone; p<0.001). This effect correlated directly with degree of MPA formation (r2 = 0.7731; p<0.0001) and was associated with increased monocyte adhesion to endothelial cells. P-selectin glycoprotein ligand-1 (PSGL-1) blocking antibody, which abrogates MPA formation, abolished these effects, as did the cyclooxygenase (COX)-2 selective inhibitor NS-398, aspirin and the EP1/EP2-selective antagonist AH6809. Conclusions/Significance These data suggest that MPA formation, as occurs in the blood under pro-inflammatory conditions, expands the pool of circulating CD14highCD16+ monocytes in a COX-2 dependent manner, and these monocytes exhibit increased adhesion to endothelium. Our findings delineate a novel mechanism underlying the pro-inflammatory effect of platelet activation.
Collapse
Affiliation(s)
- Gabriella Passacquale
- Cardiovascular Division, Department of Clinical Pharmacology, King's College London, London, United Kingdom
| | - Padman Vamadevan
- Cardiovascular Division, Department of Clinical Pharmacology, King's College London, London, United Kingdom
| | - Luis Pereira
- Cardiovascular Division, Department of Clinical Pharmacology, King's College London, London, United Kingdom
| | - Colleen Hamid
- Cardiovascular Division, Department of Clinical Pharmacology, King's College London, London, United Kingdom
| | - Valerie Corrigall
- Academic Department of Rheumatology, King's College London, London, United Kingdom
| | - Albert Ferro
- Cardiovascular Division, Department of Clinical Pharmacology, King's College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
48
|
de Mel A, Murad F, Seifalian AM. Nitric oxide: a guardian for vascular grafts? Chem Rev 2011; 111:5742-67. [PMID: 21663322 DOI: 10.1021/cr200008n] [Citation(s) in RCA: 131] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Achala de Mel
- Centre for Nanotechnology & Regenerative Medicine, Division of Surgery & Interventional Science, University College London, London, United Kingdom
| | | | | |
Collapse
|
49
|
A case of haemorrhagic cystitis by inhaled salbutamol and salmeterol. Eur J Clin Pharmacol 2011; 67:1203-4. [PMID: 21638032 DOI: 10.1007/s00228-011-1068-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Accepted: 05/20/2011] [Indexed: 10/18/2022]
|
50
|
Shah A, Passacquale G, Gkaliagkousi E, Ritter J, Ferro A. Platelet nitric oxide signalling in heart failure: role of oxidative stress. Cardiovasc Res 2011; 91:625-31. [PMID: 21502370 DOI: 10.1093/cvr/cvr115] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AIMS Heart failure is associated with deficient endothelial nitric oxide (NO) production as well as increased oxidative stress and accelerated NO degradation. The aim of this study was to evaluate platelet NO biosynthesis and superoxide anion (O(2)(-)) production in patients with heart failure. METHODS AND RESULTS In platelets from patients with heart failure due to idiopathic dilated cardiomyopathy (n= 16) and healthy control subjects (n= 23), NO synthase (NOS) activity was evaluated by L-[(3)H]-arginine to l-[(3)H]-citrulline conversion, cGMP was determined by radioimmunoassay, vasodilator-stimulated phosphoprotein (VASP: total and serine-239-phosphorylated) was assessed by western blotting, and O(2)(-) production and O(2)(-) scavenging capacity were measured by pholasin-enhanced chemiluminescence. In platelets from patients with heart failure, basal NOS activity was higher than in those from controls; furthermore, whereas platelet NOS activity increased as expected in response to albuterol or collagen in controls, no increase occurred in platelets from heart failure subjects. Despite this, basal intraplatelet NO-attributable cGMP was lower in heart failure than in control subjects, as was serine-239 phosphorylation of VASP, suggesting a decrease in bioactive NO. Platelets from heart failure subjects exhibited higher basal and collagen-stimulated O(2)(-) production and impaired O(2)(-) scavenging capacity, resulting in higher oxidative stress, consistent with the observed decrease in bioactive NO. CONCLUSION In heart failure, despite activation of NOS, platelets produce less bioactive NO, probably as a result of NO scavenging due to increased O(2)(-) production. This functional defect in the platelet l-arginine/NO/guanylyl cyclase pathway could contribute to the platelet activation observed in heart failure.
Collapse
Affiliation(s)
- Ashish Shah
- Department of Clinical Pharmacology, Cardiovascular Division, School of Medicine, King's College London, UK
| | | | | | | | | |
Collapse
|