1
|
Kozaru M, Kambara H, Higuchi A, Kagatsume T, Hosohata K. Association of Clopidogrel with Interstitial Lung Disease: Gaining Insight Through the Japanese Pharmacovigilance Database. Vasc Health Risk Manag 2024; 20:415-420. [PMID: 39247557 PMCID: PMC11378778 DOI: 10.2147/vhrm.s482190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 08/27/2024] [Indexed: 09/10/2024] Open
Abstract
Background The P2Y12 receptor inhibitors clopidogrel and prasugrel are widely used. Clopidogrel and prasugrel have different metabolic pathways, but whether their adverse event (AE) profiles differ significantly is unclear. Objective This study aimed to compare the possible AEs induced by clopidogrel and prasugrel and to assess the rank-order of their AEs submitted to a spontaneous reporting database. Materials and Methods Data were extracted from the Japanese Adverse Drug Event Report database (JADER). Reports of AEs associated with clopidogrel and prasugrel were analyzed to calculate the reporting odds ratios (RORs) and 95% confidence intervals (CIs). Results Based on 5869 reports for clopidogrel (69.6%, men) and 513 reports for prasugrel (74.1%, men), 703 and 135 different AEs were identified, respectively. Bleeding complications including hemorrhage were commonly reported for both clopidogrel and prasugrel. As for AEs related to clopidogrel, unexpected AEs such as interstitial lung disease (227 reports; ROR, 1.77; 95% CI, 1.49-2.10), abnormal hepatic function (137 reports; ROR, 1.27; 95% CI, 1.07-1.51), and hepatocellular injury (96 reports; ROR, 120.0; 95% CI, 94.9-151.8) ranked at relatively high positions based on the number of occurrences, unlike prasugrel. Conclusion This analysis of the national pharmacovigilance database highlights distinct AE profiles for clopidogrel and prasugrel. Unexpected AEs associated with clopidogrel were identified, providing valuable insights for clinical monitoring and patient safety.
Collapse
Affiliation(s)
- Mariko Kozaru
- Education and Research Center for Clinical Pharmacy, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, 569-1094, Japan
| | - Hiroko Kambara
- Education and Research Center for Clinical Pharmacy, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, 569-1094, Japan
| | - Akari Higuchi
- Education and Research Center for Clinical Pharmacy, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, 569-1094, Japan
| | - Tatsuki Kagatsume
- Education and Research Center for Clinical Pharmacy, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, 569-1094, Japan
| | - Keiko Hosohata
- Education and Research Center for Clinical Pharmacy, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka, 569-1094, Japan
| |
Collapse
|
2
|
Lother SA, Tennenhouse L, Rabbani R, Abou-Setta AM, Askin N, Turgeon AF, Murthy S, Houston BL, Houston DS, Mendelson AA, Paul JD, Farkouh ME, Hasmatali J, Rush B, Nkosi J, Goligher EC, Rimmer E, Marshall JC, Shaw SY, Lawler PR, Keynan Y, Zarychanski R. The association of antiplatelet agents with mortality among patients with non-COVID-19 community-acquired pneumonia: a systematic review and meta-analysis. Res Pract Thromb Haemost 2024; 8:102526. [PMID: 39262648 PMCID: PMC11387270 DOI: 10.1016/j.rpth.2024.102526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/16/2024] [Indexed: 09/13/2024] Open
Abstract
Background Community-acquired pneumonia (CAP) triggers inflammatory and thrombotic host responses driving morbidity and mortality. Antiplatelet agents may favorably modulate these pathways; however, their role in non-COVID-19 CAP remains uncertain. Objectives To evaluate the association of antiplatelet agents with mortality in hospitalized patients with non-COVID-19 CAP. Methods We conducted a systematic review and meta-analysis of observational studies and randomized controlled trials (RCTs) of adult patients hospitalized for non-COVID-19 CAP exposed to antiplatelet agents (acetylsalicylic acid or P2Y12 inhibitors). We searched MEDLINE, Embase, and CENTRAL from inception to August 2023. Our primary outcome was all-cause mortality: meta-analyzed (random-effects models) separately for observational studies and RCTs. For observational studies, we used adjusted mortality estimates. Results We included 13 observational studies (123,012 patients; 6 reported adjusted mortality estimates) and 2 RCTs (225 patients; both high risk of bias). In observational studies reporting hazard ratio, antiplatelet agents were associated with lower mortality (hazard ratio, 0.65; 95% CI, 0.46-0.91; I 2 = 85%; 4 studies, 91,430 patients). In studies reporting adjusted odds ratio, antiplatelet agent exposure was associated with reduced odds of mortality (odds ratio, 0.67; 95% CI, 0.45-1.00; I 2 = 0%; 2 studies, 24,889 patients). Among RCTs, there was a nonsignificant association with mortality (risk ratio, 0.66; 95% CI, 0.20-2.25; I 2 = 54%; 2 studies, 225 patients). By the Grading of Recommendations, Assessment, Development, and Evaluation criteria, the certainty of the evidence was low, primarily due to risk of bias. Conclusion In hospitalized patients with non-COVID-19 CAP, antiplatelet agents may be associated with reduced mortality compared with usual care or placebo, but the certainty of evidence is low.
Collapse
Affiliation(s)
- Sylvain A Lother
- Section of Infectious Diseases, Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- Section of Critical Care, Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Lana Tennenhouse
- Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Rasheda Rabbani
- George & Fay Yee Centre for Healthcare Innovation, Department of Community Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ahmed M Abou-Setta
- George & Fay Yee Centre for Healthcare Innovation, Department of Community Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Community Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Nicole Askin
- Neil John Maclean Health Sciences Library, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Alexis F Turgeon
- Department of Anesthesiology and Critical Care, Université Laval, Quebec City, Quebec, Canada
- Population Health and Optimal Health Practices Research Unit, Departments of Traumatology, Emergency Medicine, and Critical Care Medicine, Université Laval Research Center, Centre Hospitalier Universitaire de Quebec-Université Laval, Quebec City, Quebec, Canada
| | - Srinivas Murthy
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Brett L Houston
- Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg, Manitoba, Canada
- Section of Hematology and Medical Oncology, Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Donald S Houston
- Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg, Manitoba, Canada
- Section of Hematology and Medical Oncology, Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Asher A Mendelson
- Section of Critical Care, Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jonathan D Paul
- Section of Cardiology, Department of Medicine, University of Chicago Medical Center, Chicago, Illinois, USA
| | - Michael E Farkouh
- Department of Cardiology, Cedars-Sinai Health System, Los Angeles, California, USA
| | - Jovan Hasmatali
- Section of Critical Care, Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Barret Rush
- Section of Critical Care, Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Joel Nkosi
- Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ewan C Goligher
- Interdepartmental Division of Critical Care Medicine, and the Department of Medicine and Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Emily Rimmer
- Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg, Manitoba, Canada
- Section of Hematology and Medical Oncology, Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - John C Marshall
- Departments of Surgery and Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Souradet Y Shaw
- Department of Community Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Patrick R Lawler
- Divison of Cardiology and Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, McGill University Health Centre and McGill University, Montreal, Quebec, Canada
| | - Yoav Keynan
- Section of Infectious Diseases, Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Community Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ryan Zarychanski
- Section of Critical Care, Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Medical Oncology and Hematology, CancerCare Manitoba, Winnipeg, Manitoba, Canada
- Section of Hematology and Medical Oncology, Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
3
|
Parker WAE, Storey RF. The role of platelet P2Y 12 receptors in inflammation. Br J Pharmacol 2024; 181:515-531. [PMID: 37771103 DOI: 10.1111/bph.16256] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 08/15/2023] [Accepted: 09/15/2023] [Indexed: 09/30/2023] Open
Abstract
Inflammation is a complex pathophysiological process underlying many clinical conditions. Platelets contribute to the thrombo-inflammatory response. Platelet P2Y12 receptors amplify platelet activation, potentiating platelet aggregation, degranulation and shape change. The contents of platelet alpha granules, in particular, act directly on leucocytes, including mediating platelet-leucocyte aggregation and activation via platelet P-selectin. Much evidence for the role of platelet P2Y12 receptors in inflammation comes from studies using antagonists of these receptors, such as the thienopyridines clopidogrel and prasugrel, and the cyclopentyltriazolopyrimidine ticagrelor, in animal and human experimental models. These suggest that antagonism of P2Y12 receptors decreases markers of inflammation with some evidence that this reduces incidence of adverse clinical sequelae during inflammatory conditions. Interpretation is complicated by pleiotropic effects such as those of the thienopyridines on circulating leucocyte numbers and of ticagrelor on adenosine reuptake. The available evidence suggests that P2Y12 receptors are prominent mediators of inflammation and P2Y12 receptor antagonism as a potentially powerful strategy in a broad range of inflammatory conditions. LINKED ARTICLES: This article is part of a themed issue on Platelet purinergic receptor and non-thrombotic disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.4/issuetoc.
Collapse
Affiliation(s)
- William A E Parker
- Cardiovascular Research Unit, Division of Clinical Medicine, University of Sheffield, Sheffield, UK
- NIHR Sheffield Biomedical Research Centre, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Robert F Storey
- Cardiovascular Research Unit, Division of Clinical Medicine, University of Sheffield, Sheffield, UK
- NIHR Sheffield Biomedical Research Centre, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| |
Collapse
|
4
|
Yeh JS, Chen WT, Tomlinson B, Tam WC, Chien LN. Comparing the effectiveness and safety of dual antiplatelet with ticagrelor or clopidogrel in elderly Asian patients with acute myocardial infraction. Front Cardiovasc Med 2023; 10:1143509. [PMID: 37008324 PMCID: PMC10060791 DOI: 10.3389/fcvm.2023.1143509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/27/2023] [Indexed: 03/17/2023] Open
Abstract
BackgroundCurrent guidelines recommend potent P2Y12 inhibitors for patients after acute coronary syndrome. However, the data on the efficacy and safety of potent P2Y12 inhibitors in elderly Asian populations was limited. We aimed to investigate the major adverse cardiovascular events (MACE), bleeding events, and net adverse clinical events (NACE) with ticagrelor and clopidogrel in Taiwanese patients aged 65 and older after acute myocardial infarction (AMI).MethodsThis retrospective population-based cohort study was conducted using data from the National Health Insurance Research Database. The AMI patients aged ≥65 years who underwent percutaneous coronary intervention (PCI) and survived after 1 month were included. The patients were separated into 2 cohorts depending on the type of dual antiplatelet therapy (DAPT) they received: ticagrelor plus aspirin (T + A) or clopidogrel plus aspirin (C + A). We used inverse probability of treatment weighting to balance the difference between these 2 study groups. The outcome included all-cause mortality, MACE (cardiovascular death, nonfatal ischemic stroke, and nonfatal myocardial infarction), intracerebral hemorrhage, major bleeding, and NACE which is composed of cardiovascular death, ischemic and hemorrhagic events. The follow-up period was up to 12 months.ResultsFrom 2013 to 2017, a total of 14,715 patients who met the eligibility criteria were separated into 2 groups: 5,051 for T + A and 9,664 for C + A. Compared to patients with C + A, patients who received T + A had a lower risk of cardiovascular death and all-cause death, with an adjusted HR of 0.57 [95% confidence interval (CI), 0.38–0.85, p = 0.006] and 0.58 (95% CI 0.45–0.74, p < 0.001), respectively. No differences were found in MACE, intracranial and major bleeding between the 2 groups. In addition, the patients with T + A had a lower risk of NACE with an adjusted HR of 0.86 (95% CI 0.74–1.00, p = 0.045)ConclusionAmong elderly AMI patients receiving DAPT after successful PCI, ticagrelor was a more favorable P2Y12 inhibitor than clopidogrel because of lowering the risk of death and NACE without increasing the risk of severe bleeding. Ticagrelor is an effective and safe P2Y12 inhibitor in Asian elderly survivors after PCI.
Collapse
Affiliation(s)
- Jong-Shiuan Yeh
- Division of Cardiovascular Medicine, Department of Internal Medicine, Taipei Municipal Wan-Fang Hospital, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wan-Ting Chen
- Health Data Analytics and Statistics Center, Office of Data Science, Taipei Medical University, Taipei, Taiwan
| | - Brian Tomlinson
- Faculty of Medicine, Macau University of Science and Technology, Macao SAR, China
| | - Weng-Chio Tam
- Department of Cardiology, Centro Hospitalar Conde São Januário, Macao SAR, China
| | - Li-Nien Chien
- Institute of Health and Welfare Policy, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Graduate Institute of Data Science, College of Management, Taipei Medical University, Taipei, Taiwan
- Correspondence: Li-Nien Chien
| |
Collapse
|
5
|
Gentry CA, Williams RJ, Whitman CM, Thind SK, Kliewer BS. Staphylococcus aureus bacteraemia treatment outcomes in patients receiving ticagrelor vs a propensity-matched cohort receiving clopidogrel. Int J Antimicrob Agents 2023; 61:106743. [PMID: 36736927 DOI: 10.1016/j.ijantimicag.2023.106743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023]
Abstract
OBJECTIVES Ticagrelor may improve the outcomes in Staphylococcus aureus bacteraemia (SAB). However, treatment outcome data for these patients remain limited. The primary objective of this study was to characterize the outcomes of patients with SAB who received ticagrelor compared with a cohort who received clopidogrel. METHODS This was a retrospective, nationwide propensity-matched analysis of patients with SAB who were prescribed ticagrelor or clopidogrel concomitantly with antistaphylococcal therapy. The primary outcome was the comparative all-cause 30-day mortality rate between propensity-matched groups. RESULTS In total, 1509 patients were prescribed concomitantly with ticagrelor or clopidogrel during treatment of S. aureus bacteraemia; of these, 194 patients were excluded from this study due to an inadequate number of antiplatelet doses within the first week of therapy (n=171) or non-admission to hospital (n=23). Of the remaining 1315 patients, 74 patients received ticagrelor and 1241 patients received clopidogrel. There was no significant difference in all-cause 30-day mortality between the groups [6/74 (8.1%) in the ticagrelor group vs 10/74 (13.5%) in the clopidogrel group; P=0.29]. Multi-variate logistic regression demonstrated that elevated aspartate aminotransferase, systolic blood pressure <90 mmHg, elevated serum creatinine and neurological comorbidity were independently associated with all-cause 30-day mortality. CONCLUSIONS This study found no difference in all-cause 30-day mortality between the two groups, although overall mortality appeared to be lower compared with other reports. Randomized controlled trials of P2Y12 inhibitors as adjunctive agents to antibiotic therapy for the treatment of serious S. aureus infections are warranted.
Collapse
Affiliation(s)
- Chris A Gentry
- Pharmacy Service, Oklahoma City Veterans Affairs Health Care System, Oklahoma City, Oklahoma, USA.
| | - Riley J Williams
- Pharmacy Service, Oklahoma City Veterans Affairs Health Care System, Oklahoma City, Oklahoma, USA
| | - Charles M Whitman
- Pharmacy Service, Oklahoma City Veterans Affairs Health Care System, Oklahoma City, Oklahoma, USA
| | - Sharanjeet K Thind
- Section of Infectious Diseases, Medical Service, Oklahoma City Veterans Affairs Health Care System, Oklahoma City, Oklahoma, USA; Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Brian S Kliewer
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA; Section of Internal Medicine, Medical Service, Oklahoma City Veterans Affairs Health Care System, Oklahoma City, Oklahoma, USA
| |
Collapse
|
6
|
Minasyan H. Oxygen therapy for sepsis and prevention of complications. Acute Crit Care 2022; 37:137-150. [PMID: 35545238 PMCID: PMC9184979 DOI: 10.4266/acc.2021.01200] [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: 08/13/2021] [Accepted: 12/16/2021] [Indexed: 11/30/2022] Open
Abstract
Patients with sepsis have a wide range of respiratory disorders that can be treated with oxygen therapy. Experimental data in animal sepsis models show that oxygen therapy significantly increases survival, while clinical data on the use of different oxygen therapy protocols are ambiguous. Oxygen therapy, especially hyperbaric oxygenation, in patients with sepsis can aggravate existing oxidative stress and contribute to the development of disseminated intravascular coagulation. The purpose of this article is to compare experimental and clinical data on oxygen therapy in animals and humans, to discuss factors that can influence the results of oxygen therapy for sepsis treatment in humans, and to provide some recommendations for reducing oxidative stress and preventing disseminated intravascular coagulation during oxygen therapy.
Collapse
|
7
|
Rabouël Y, Magnenat S, Lefebvre F, Delabranche X, Gachet C, Hechler B. Transfusion of fresh washed platelets does not prevent experimental polymicrobial-induced septic shock in mice. J Thromb Haemost 2022; 20:449-460. [PMID: 34752015 DOI: 10.1111/jth.15583] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 11/29/2022]
Abstract
INTRODUCTION The specific role of platelets during sepsis is not yet fully understood, probably related to the paradox of platelets being potentially beneficial but also deleterious via their thrombotic functions. OBJECTIVE To evaluate the impact of thrombocytopenia on septic shock in mice and to investigate whether transfusion of fresh washed platelets, either fully functional or with impaired hemostatic properties, might have beneficial effects. METHODS Septic shock was induced by cecal ligation and puncture (CLP). Experimental depletion of circulating platelets was induced with a rat anti-mouse GPIbα monoclonal antibody. Transfusion of either wild-type washed platelets, platelets treated with the antiplatelet drugs acetylsalicylic acid (ASA) and clopidogrel, or GPIIbIIIa-deficient washed platelets treated with ASA and clopidogrel was performed 4 h after CLP surgery. RESULTS Depletion of circulating platelets negatively affected septic shock, worsening systemic inflammation, coagulopathy, organ damage, and mortality, raising the question of whether a higher platelet count could be protective. Transfusion of fully functional platelets or platelets with combined treatment with ASA and clopidogrel, with or without additional GPIIbIIIa deficiency, afforded an immediate return of circulating platelet counts to their initial values before surgery. However, transfusion of each of the three types of platelets did not prevent arterial hypotension, inflammatory response, coagulopathy, and organ damage during septic shock. CONCLUSION Depletion of circulating platelets negatively affects septic shock, while transfusion of washed platelets has no significant beneficial effect in mice.
Collapse
Affiliation(s)
- Yannick Rabouël
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand Est, BPPS UMR_S1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Stéphanie Magnenat
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand Est, BPPS UMR_S1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Floryna Lefebvre
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand Est, BPPS UMR_S1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Xavier Delabranche
- Hôpitaux Universitaires de Strasbourg, Anesthésie, Réanimation et Médecine périopératoire, Nouvel Hôpital Civil, Strasbourg, France
| | - Christian Gachet
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand Est, BPPS UMR_S1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Beatrice Hechler
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand Est, BPPS UMR_S1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| |
Collapse
|
8
|
Vardon-Bounes F, Garcia C, Piton A, Series J, Gratacap MP, Poëtte M, Seguin T, Crognier L, Ruiz S, Silva S, Conil JM, Minville V, Payrastre B. Evolution of Platelet Activation Parameters During Septic Shock in Intensive Care Unit. Platelets 2021; 33:918-925. [PMID: 34915822 DOI: 10.1080/09537104.2021.2007873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
During severe sepsis, platelet activation may induce disseminate microvascular thrombosis, which play a key role in critical organ failure. Crucially, most of the studies in this field have explored platelet-leukocyte interactions in animal models, or explored platelets under the spectrum of thrombocytopenia or disseminated intravascular coagulation and have not taken into account the complex interplay that might exist between platelets and leukocytes during human septic shock nor the kinetics of platelet activation. Here, we assessed platelet activation parameters at the admission of patients with sepsis to the intensive care unit (ICU) and 48 hours later. Twenty-two patients were enrolled in the study, thirteen (59.1%) of whom were thrombocytopenic. The control group was composed of twelve infection-free patients admitted during the study period. The activation parameters studied included platelet-leukocyte interactions, assessed by flow cytometry in whole blood, as well as membrane surface and soluble platelet activation markers measured by flow cytometry and dedicated ELISA kits. We also investigated platelet aggregation and secretion responses of patients with sepsis following stimulation, compared to controls. At admission, the level of circulating monocyte-platelet and neutrophil-platelet heterotypic aggregates was significantly higher in sepsis patients compared to controls and returned to a level comparable to controls or even below 48 hours later. Basal levels of CD62P and CD63 platelet membrane exposure at admission and 48 hours later were low and similar to controls. In contrast, plasma level of soluble GPVI and soluble CD40 ligand was significantly increased in septic patients, at the two times of analysis, reflecting previous platelet activation. Platelet aggregation and secretion responses induced by specific agonists were significantly decreased in septic conditions, particularly 48 hours after admission. Hence, we have observed for the first time that critically ill septic patients compared to controls have both an early and durable platelet activation while their circulating platelets are less responsive to different agonists.
Collapse
Affiliation(s)
- Fanny Vardon-Bounes
- Pôle Anesthésie-Réanimation, Centre Hospitalier Universitaire de Toulouse, Toulouse, France.,INSERM UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université Paul Sabatier Toulouse 3, Toulouse, France
| | - Cédric Garcia
- INSERM UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université Paul Sabatier Toulouse 3, Toulouse, France.,Laboratoire d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Alexandra Piton
- Pôle Anesthésie-Réanimation, Centre Hospitalier Universitaire de Toulouse, Toulouse, France.,INSERM UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université Paul Sabatier Toulouse 3, Toulouse, France
| | - Jennifer Series
- INSERM UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université Paul Sabatier Toulouse 3, Toulouse, France.,Laboratoire d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Marie-Pierre Gratacap
- INSERM UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université Paul Sabatier Toulouse 3, Toulouse, France.,Laboratoire d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Michaël Poëtte
- Pôle Anesthésie-Réanimation, Centre Hospitalier Universitaire de Toulouse, Toulouse, France.,INSERM UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université Paul Sabatier Toulouse 3, Toulouse, France
| | - Thierry Seguin
- Pôle Anesthésie-Réanimation, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Laure Crognier
- Pôle Anesthésie-Réanimation, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Stéphanie Ruiz
- Pôle Anesthésie-Réanimation, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Stein Silva
- Pôle Anesthésie-Réanimation, Centre Hospitalier Universitaire de Toulouse, Toulouse, France.,INSERM UMR 1214, ToNIC: Toulouse NeuroImaging Center, Toulouse, France
| | - Jean-Marie Conil
- Pôle Anesthésie-Réanimation, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Vincent Minville
- Pôle Anesthésie-Réanimation, Centre Hospitalier Universitaire de Toulouse, Toulouse, France.,INSERM UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université Paul Sabatier Toulouse 3, Toulouse, France
| | - Bernard Payrastre
- INSERM UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université Paul Sabatier Toulouse 3, Toulouse, France.,Laboratoire d'Hématologie, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| |
Collapse
|
9
|
Hu Y, Yu C, Guo Y, Bian Z, Han Y, Yang L, Chen Y, Du H, Pang Y, Sun D, Jin J, Zhang J, Wang J, Shao C, Tang YD, Chen J, Chen Z, Lv J, Li L. Pneumonia hospitalizations and the subsequent risk of incident ischaemic cardiovascular disease in Chinese adults. Int J Epidemiol 2021; 50:1698-1707. [PMID: 33826715 DOI: 10.1093/ije/dyab039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 02/16/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Acute respiratory infections have been associated with a transient increase in cardiovascular risk. However, whether such an association persists beyond 1 month and the potential modifying effect of cardiovascular risk factors on such an association are less well established. METHODS The China Kadoorie Biobank enrolled 512 726 participants aged 30-79 years from 10 areas across China during 2004-2008. By the end of 2017, a total of 5444 participants with new-onset ischaemic heart disease (IHD) and 4846 with ischaemic stroke (IS) who also had at least a record of hospitalization for pneumonia during follow-up were included. We used a self-controlled case-series method and calculated the age- and season-adjusted relative incidences (RIs) and 95% confidence intervals (CIs) for ischaemic cardiovascular disease (CVD) after pneumonia. RESULTS The risk of ischaemic CVD increased during days 1-3 after pneumonia hospitalization, with an RI (95% CI) of 4.24 (2.92-6.15) for IHD and 1.85 (1.02-3.35) for IS. The risk gradually reduced with longer duration since pneumonia hospitalization but remained elevated until days 92-365 for IHD (1.23, 1.12-1.35) and days 29-91 for IS (1.25, 1.05-1.48). Pre-existing cardiovascular risk factors amplified the associations between pneumonia and ischaemic CVD risks, such as chronic obstructive pulmonary disease for both IHD and IS, and diabetes and smoking for IHD (all Pinteraction < 0.05). Besides, the risk of ischaemic CVD was also higher among the participants aged ≥70 years (Pinteraction < 0.001 for IHD and 0.033 for IS). CONCLUSION Among middle-aged and older Chinese adults, pneumonia hospitalization was associated with both short- and long-term increases in ischaemic CVD risk for ≤1 year.
Collapse
Affiliation(s)
- Yizhen Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
| | - Yu Guo
- Chinese Academy of Medical Sciences, Beijing, China
| | - Zheng Bian
- Chinese Academy of Medical Sciences, Beijing, China
| | - Yuting Han
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Ling Yang
- Medical Research Council Population Health Research Unit at the University of Oxford, Oxford, UK
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Yiping Chen
- Medical Research Council Population Health Research Unit at the University of Oxford, Oxford, UK
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Huaidong Du
- Medical Research Council Population Health Research Unit at the University of Oxford, Oxford, UK
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Yuanjie Pang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Dianjianyi Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Jianrong Jin
- Wuzhong District Center for Disease Control and Prevention, Jiangsu, China
| | - Jun Zhang
- Suzhou Center for Disease Control and Prevention, Jiangsu, China
| | - Jingjia Wang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunli Shao
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi-Da Tang
- Department of Cardiology, Third Hospital, Peking University, Beijing, China
| | - Junshi Chen
- China National Center for Food Safety Risk Assessment, Beijing, China
| | - Zhengming Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences (Peking University), Ministry of Education, Beijing, China
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
| |
Collapse
|
10
|
Vicent L, Bruña V, Devesa C, Sousa-Casasnovas I, Juárez M, Alcalá L, Muñoz P, Fernández-Avilés F, Martínez-Sellés M. Ticagrelor and Infection Risk in Patients with Coronary Artery Disease. Cardiology 2021; 146:698-704. [PMID: 34551409 DOI: 10.1159/000519285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 08/27/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Ticagrelor has a bactericidal effect in vitro, and clinical studies suggest a beneficial effect in infections. Our aim was to determine the incidence of infections in patients treated with 3 different P2Y12 receptor inhibitors. METHODS Retrospective registry in a cardiology department. Patients with coronary artery disease discharged on ticagrelor, prasugrel, or clopidogrel from March 2017 to June 2019 were included. The risk of infection was analyzed during the period of P2Y12 inhibitor treatment (12.4 ± 6.7 months). RESULTS A total of 250 patients were included (ticagrelor 91 [36.4%], prasugrel 89 [35.6%], clopidogrel 70 [28.0%]). Mean age was 61.0 ± 13.1 years, and 63 (25.2%) were women. The most common reason to use these drugs was ST-segment elevation acute myocardial infarction (STEMI) (152 patients - 60.8%). STEMI was the reason to use prasugrel in 84 patients (94.4%), ticagrelor in 44 (48.4%), and clopidogrel in 24 (34.3%), p < 0.001. An infection during follow-up was seen in 87 patients (34.8%), 23 treated with ticagrelor (25.3%), 30 with prasugrel (33.7%) and 34 with clopidogrel (48.6%), p = 0.009. Ticagrelor was independently associated with a lower likelihood of infection (Hazard Ratio [HR] 0.52, 95% confidence interval [CI] 0.28-0.95; p = 0.035) compared to prasugrel (HR 0.96, 95% CI 0.54-1.73; p = 0.909) and clopidogrel (HR = 1). CONCLUSIONS In patients admitted with coronary artery disease patients treated with ticagrelor had a lower frequency of infections during follow-up than those treated with other P2Y12 inhibitors. Further studies are necessary to clarify the bactericidal effect of ticagrelor in this context.
Collapse
Affiliation(s)
- Lourdes Vicent
- Servicio de Cardiología, Hospital General Universitario 12 de Octubre, Madrid, Spain, .,CIBER Enfermedades Cardiovasculares - CIBERCV, Madrid, Spain,
| | - Vanesa Bruña
- Servicio de Cardiología, Hospital General Universitario 12 de Octubre, Madrid, Spain.,CIBER Enfermedades Cardiovasculares - CIBERCV, Madrid, Spain
| | - Carolina Devesa
- CIBER Enfermedades Cardiovasculares - CIBERCV, Madrid, Spain.,Servicio de Cardiología, Hospital General Universitario Gregorio Marañón, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital Gregorio Marañón, Madrid, Spain
| | - Iago Sousa-Casasnovas
- CIBER Enfermedades Cardiovasculares - CIBERCV, Madrid, Spain.,Servicio de Cardiología, Hospital General Universitario Gregorio Marañón, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital Gregorio Marañón, Madrid, Spain
| | - Miriam Juárez
- CIBER Enfermedades Cardiovasculares - CIBERCV, Madrid, Spain.,Servicio de Cardiología, Hospital General Universitario Gregorio Marañón, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital Gregorio Marañón, Madrid, Spain
| | - Luis Alcalá
- Instituto de Investigación Sanitaria Hospital Gregorio Marañón, Madrid, Spain.,Servicio de Microbiología Clínica y Enfermedades Infecciosas, Hospital General Universitario Gregorio Marañón, Madrid, Spain.,CIBER Enfermedades Respiratorias- CIBERES (CB06/06/0058), Madrid, Spain
| | - Patricia Muñoz
- Instituto de Investigación Sanitaria Hospital Gregorio Marañón, Madrid, Spain.,Servicio de Microbiología Clínica y Enfermedades Infecciosas, Hospital General Universitario Gregorio Marañón, Madrid, Spain.,CIBER Enfermedades Respiratorias- CIBERES (CB06/06/0058), Madrid, Spain.,Universidad Complutense, Madrid, Spain
| | - Francisco Fernández-Avilés
- CIBER Enfermedades Cardiovasculares - CIBERCV, Madrid, Spain.,Servicio de Cardiología, Hospital General Universitario Gregorio Marañón, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital Gregorio Marañón, Madrid, Spain.,Universidad Complutense, Madrid, Spain
| | - Manuel Martínez-Sellés
- CIBER Enfermedades Cardiovasculares - CIBERCV, Madrid, Spain.,Servicio de Cardiología, Hospital General Universitario Gregorio Marañón, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital Gregorio Marañón, Madrid, Spain.,Universidad Complutense, Madrid, Spain.,Universidad Europea, Madrid, Spain
| |
Collapse
|
11
|
Rabouel Y, Magnenat S, Delabranche X, Gachet C, Hechler B. Platelet P2Y 12 Receptor Deletion or Pharmacological Inhibition does not Protect Mice from Sepsis or Septic Shock. TH OPEN 2021; 5:e343-e352. [PMID: 34447900 PMCID: PMC8384481 DOI: 10.1055/s-0041-1733857] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 07/06/2021] [Indexed: 12/27/2022] Open
Abstract
Introduction
Platelets are increasingly appreciated as key effectors during sepsis, raising the question of the usefulness of antiplatelet drugs to treat patients with sepsis.
Objective
Evaluate the potential contribution of the platelet P2Y
12
receptor in the pathogenesis of polymicrobial-induced sepsis and septic shock in mice.
Methods
The effects of P2Y
12
inhibition using clopidogrel treatment and of platelet-specific deletion of the P2Y
12
receptor in mice were examined in two severity grades of cecal ligation and puncture (CLP) leading to mild sepsis or septic shock.
Results
Twenty hours after induction of the high grade CLP, clopidogrel- and vehicle-treated mice displayed a similar 30% decrease in mean arterial blood pressure (MAP) characteristic of shock. Septic shock-induced thrombocytopenia was not modified by clopidogrel treatment. Plasma concentrations of inflammatory cytokines and myeloperoxidase (MPO) were similarly increased in clopidogrel- and vehicle-treated mice, indicating comparable increase in systemic inflammation. Thrombin-antithrombin (TAT) complexes and the extent of organ damage were also similar. In mild-grade CLP, clopidogrel- and vehicle-treated mice did not display a significant decrease in MAP, while thrombocytopenia and plasma concentrations of TNFα, IL6, IL10, MPO, TAT and organ damage reached similar levels in both groups, although lower than those reached in the high grade CLP. Similarly, mice with platelet-specific deletion of the P2Y
12
receptor were not protected from CLP-induced sepsis or septic shock.
Conclusion
The platelet P2Y
12
receptor does not contribute to the pathogenesis of sepsis or septic shock in mice, suggesting that P2Y
12
receptor antagonists may not be beneficial in patients with sepsis or septic shock.
Collapse
Affiliation(s)
- Yannick Rabouel
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS)-Grand Est, BPPS UMR_S 1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), F-67000 Strasbourg, France
| | - Stéphanie Magnenat
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS)-Grand Est, BPPS UMR_S 1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), F-67000 Strasbourg, France
| | - Xavier Delabranche
- Hôpitaux Universitaires de Strasbourg, Anesthésie, Réanimation et Médecine périopératoire, Nouvel Hôpital Civil, F-67000 Strasbourg, France
| | - Christian Gachet
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS)-Grand Est, BPPS UMR_S 1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), F-67000 Strasbourg, France
| | - Beatrice Hechler
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS)-Grand Est, BPPS UMR_S 1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), F-67000 Strasbourg, France
| |
Collapse
|
12
|
Sun J, Uchiyama S, Olson J, Morodomi Y, Cornax I, Ando N, Kohno Y, Kyaw MMT, Aguilar B, Haste NM, Kanaji S, Kanaji T, Rose WE, Sakoulas G, Marth JD, Nizet V. Repurposed drugs block toxin-driven platelet clearance by the hepatic Ashwell-Morell receptor to clear Staphylococcus aureus bacteremia. Sci Transl Med 2021; 13:13/586/eabd6737. [PMID: 33762439 DOI: 10.1126/scitranslmed.abd6737] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 03/01/2021] [Indexed: 12/13/2022]
Abstract
Staphylococcus aureus (SA) bloodstream infections cause high morbidity and mortality (20 to 30%) despite modern supportive care. In a human bacteremia cohort, we found that development of thrombocytopenia was correlated to increased mortality and increased α-toxin expression by the pathogen. Platelet-derived antibacterial peptides are important in bloodstream defense against SA, but α-toxin decreased platelet viability, induced platelet sialidase to cause desialylation of platelet glycoproteins, and accelerated platelet clearance by the hepatic Ashwell-Morell receptor (AMR). Ticagrelor (Brilinta), a commonly prescribed P2Y12 receptor inhibitor used after myocardial infarction, blocked α-toxin-mediated platelet injury and resulting thrombocytopenia, thereby providing protection from lethal SA infection in a murine intravenous challenge model. Genetic deletion or pharmacological inhibition of AMR stabilized platelet counts and enhanced resistance to SA infection, and the anti-influenza sialidase inhibitor oseltamivir (Tamiflu) provided similar therapeutic benefit. Thus, a "toxin-platelet-AMR" regulatory pathway plays a critical role in the pathogenesis of SA bloodstream infection, and its elucidation provides proof of concept for repurposing two commonly prescribed drugs as adjunctive therapies to improve patient outcomes.
Collapse
Affiliation(s)
- Josh Sun
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA.,Department of Pediatrics, UC San Diego, La Jolla, CA 92093, USA.,Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, CA 92093, USA
| | - Satoshi Uchiyama
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA
| | - Joshua Olson
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA
| | - Yosuke Morodomi
- Department of Molecular Medicine, MERU-Roon Research Center on Vascular Biology, Scripps Research, La Jolla, CA 92037, USA
| | - Ingrid Cornax
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA
| | - Nao Ando
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA
| | - Yohei Kohno
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA
| | - May M T Kyaw
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA
| | - Bernice Aguilar
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA
| | - Nina M Haste
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA.,Department of Pediatrics, UC San Diego, La Jolla, CA 92093, USA.,Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, CA 92093, USA
| | - Sachiko Kanaji
- Department of Molecular Medicine, MERU-Roon Research Center on Vascular Biology, Scripps Research, La Jolla, CA 92037, USA
| | - Taisuke Kanaji
- Department of Molecular Medicine, MERU-Roon Research Center on Vascular Biology, Scripps Research, La Jolla, CA 92037, USA
| | - Warren E Rose
- School of Pharmacy, University of Wisconsin, Madison, WI 53705, USA
| | - George Sakoulas
- Department of Pediatrics, UC San Diego, La Jolla, CA 92093, USA
| | - Jamey D Marth
- Center for Nanomedicine, UC Santa Barbara, Santa Barbara, CA 93106, USA.,Sanford Burnham Prebys Medical Discovery Institute, UC Santa Barbara, Santa Barbara, CA 93106, USA
| | - Victor Nizet
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA. .,Department of Pediatrics, UC San Diego, La Jolla, CA 92093, USA.,Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
13
|
Luo Q, Liu R, Qu K, Liu G, Hang M, Chen G, Xu L, Jin Q, Guo D, Kang Q. Cangrelor ameliorates CLP-induced pulmonary injury in sepsis by inhibiting GPR17. Eur J Med Res 2021; 26:70. [PMID: 34229761 PMCID: PMC8262027 DOI: 10.1186/s40001-021-00536-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 06/21/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Sepsis is a common complication of severe wound injury and infection, with a very high mortality rate. The P2Y12 receptor inhibitor, cangrelor, is an antagonist anti-platelet drug. METHODS In our study, we investigated the protective mechanisms of cangrelor in CLP-induced pulmonary injury in sepsis, using C57BL/6 mouse models. RESULTS TdT-mediated dUTP Nick-End Labeling (TUNEL) and Masson staining showed that apoptosis and fibrosis in lungs were alleviated by cangrelor treatment. Cangrelor significantly promoted surface expression of CD40L on platelets and inhibited CLP-induced neutrophils in Bronchoalveolar lavage fluid (BALF) (p < 0.001). We also found that cangrelor decreased the inflammatory response in the CLP mouse model and inhibited the expression of inflammatory cytokines, IL-1β (p < 0.01), IL-6 (p < 0.05), and TNF-α (p < 0.001). Western blotting and RT-PCR showed that cangrelor inhibited the increased levels of G-protein-coupled receptor 17 (GPR17) induced by CLP (p < 0.001). CONCLUSION Our study indicated that cangrelor repressed the levels of GPR17, followed by a decrease in the inflammatory response and a rise of neutrophils in BALF, potentially reversing CLP-mediated pulmonary injury during sepsis.
Collapse
Affiliation(s)
- Qiancheng Luo
- Department of Critical Care Medicine, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People's Republic of China
| | - Rui Liu
- Department of Endocrinology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People's Republic of China
| | - Kaili Qu
- Postgraduate Training Base in Shanghai Gongli Hospital, Ningxia Medical University, Shanghai, 200135, People's Republic of China
| | - Guorong Liu
- Department of Critical Care Medicine, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People's Republic of China
| | - Min Hang
- Department of Critical Care Medicine, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People's Republic of China
| | - Guo Chen
- Department of Critical Care Medicine, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People's Republic of China
| | - Lei Xu
- Department of Critical Care Medicine, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People's Republic of China
| | - Qinqin Jin
- Department of Critical Care Medicine, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People's Republic of China
| | - Dongfeng Guo
- Department of Critical Care Medicine, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People's Republic of China.
| | - Qi Kang
- Department of Critical Care Medicine, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, 200135, People's Republic of China.
| |
Collapse
|
14
|
Morris G, Bortolasci CC, Puri BK, Olive L, Marx W, O'Neil A, Athan E, Carvalho A, Maes M, Walder K, Berk M. Preventing the development of severe COVID-19 by modifying immunothrombosis. Life Sci 2021; 264:118617. [PMID: 33096114 PMCID: PMC7574725 DOI: 10.1016/j.lfs.2020.118617] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 10/01/2020] [Accepted: 10/13/2020] [Indexed: 01/10/2023]
Abstract
BACKGROUND COVID-19-associated acute respiratory distress syndrome (ARDS) is associated with significant morbidity and high levels of mortality. This paper describes the processes involved in the pathophysiology of COVID-19 from the initial infection and subsequent destruction of type II alveolar epithelial cells by SARS-CoV-2 and culminating in the development of ARDS. MAIN BODY The activation of alveolar cells and alveolar macrophages leads to the release of large quantities of proinflammatory cytokines and chemokines and their translocation into the pulmonary vasculature. The presence of these inflammatory mediators in the vascular compartment leads to the activation of vascular endothelial cells platelets and neutrophils and the subsequent formation of platelet neutrophil complexes. These complexes in concert with activated endothelial cells interact to create a state of immunothrombosis. The consequence of immunothrombosis include hypercoagulation, accelerating inflammation, fibrin deposition, migration of neutrophil extracellular traps (NETs) producing neutrophils into the alveolar apace, activation of the NLRP3 inflammazome, increased alveolar macrophage destruction and massive tissue damage by pyroptosis and necroptosis Therapeutic combinations aimed at ameliorating immunothrombosis and preventing the development of severe COVID-19 are discussed in detail.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Chiara C Bortolasci
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, Centre for Molecular and Medical Research, School of Medicine, Geelong, Australia
| | | | - Lisa Olive
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; School of Psychology, Deakin University, Geelong, Australia
| | - Wolfgang Marx
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Adrienne O'Neil
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Melbourne School of Population and Global Health, Melbourne, Australia
| | - Eugene Athan
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Barwon Health, Geelong, Australia
| | - Andre Carvalho
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, University of Toronto, Toronto, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, Canada
| | - Michael Maes
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, King Chulalongkorn University Hospital, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Ken Walder
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, Centre for Molecular and Medical Research, School of Medicine, Geelong, Australia
| | - Michael Berk
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, Centre for Youth Mental Health, Florey Institute for Neuroscience and Mental Health and the Department of Psychiatry, The University of Melbourne, Melbourne, Australia.
| |
Collapse
|
15
|
Ghimire S, Ravi S, Budhathoki R, Arjyal L, Hamal S, Bista A, Khadka S, Uprety D. Current understanding and future implications of sepsis-induced thrombocytopenia. Eur J Haematol 2020; 106:301-305. [PMID: 33191517 DOI: 10.1111/ejh.13549] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023]
Abstract
Sepsis is a global health burden that needs intensive medical care. Thrombocytopenia in sepsis is well known to increase morbidity as well as mortality. Several studies have been performed both in animal models and in humans to understand the mechanism by which sepsis causes thrombocytopenia. Recent studies have shown that inhibiting thrombocytopenia improves outcomes in sepsis patients. Understanding these mechanisms to identify targets in use of newer treatment modalities besides using resuscitation measures, antibiotics and removal of thrombocytopenia inducing agent could potentially help us improve outcomes in sepsis.
Collapse
Affiliation(s)
- Subash Ghimire
- Department of Medicine, Guthrie Robert Packer Hospital, Sayre, PA, USA
| | - Swapna Ravi
- Department of Medicine, Guthrie Robert Packer Hospital, Sayre, PA, USA
| | - Rasmita Budhathoki
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Lubina Arjyal
- Department of Medicine, Gundersen Lutheran Medical Center, La Crosse, WI, USA
| | - Savyata Hamal
- Zainul Haque Sikder Women's Medical College, Bangladesh
| | - Amir Bista
- Department of Hematology-Oncology, Gundersen Lutheran Medical Center, LA Crosse, WI, USA
| | - Sushmita Khadka
- Department of Medicine, Guthrie Robert Packer Hospital, Sayre, PA, USA
| | - Dipesh Uprety
- Department of Hematology-Oncology, Gundersen Lutheran Medical Center, LA Crosse, WI, USA
| |
Collapse
|
16
|
P2Y 12 Inhibition beyond Thrombosis: Effects on Inflammation. Int J Mol Sci 2020; 21:ijms21041391. [PMID: 32092903 PMCID: PMC7073040 DOI: 10.3390/ijms21041391] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/14/2020] [Accepted: 02/15/2020] [Indexed: 12/18/2022] Open
Abstract
The P2Y12 receptor is a key player in platelet activation and a major target for antithrombotic drugs. The beneficial effects of P2Y12 receptor antagonists might, however, not be restricted to the primary and secondary prevention of arterial thrombosis. Indeed, it has been established that platelet activation also has an essential role in inflammation. Additionally, nonplatelet P2Y12 receptors present in immune cells and vascular smooth muscle cells might be effective players in the inflammatory response. This review will investigate the biological and clinical impact of P2Y12 receptor inhibition beyond its platelet-driven antithrombotic effects, focusing on its anti-inflammatory role. We will discuss the potential molecular and cellular mechanisms of P2Y12-mediated inflammation, including cytokine release, platelet–leukocyte interactions and neutrophil extracellular trap formation. Then we will summarize the current evidence on the beneficial effects of P2Y12 antagonists during various clinical inflammatory diseases, especially during sepsis, acute lung injury, asthma, atherosclerosis, and cancer.
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW Pneumonia, an inflammatory disease, is the single largest infectious cause of death. Pneumonia has recently been established as an important contributing factor to major adverse cardiovascular events including heart failure. Developing an intermechanistic understanding of pneumonia and cardiovascular disease is crucial for successful future drug therapy and reducing healthcare expenditure. RECENT FINDINGS Up to 30% of patients admitted with pneumonia develop cardiovascular complications such as heart failure within 10 years of hospital discharge. Recent mechanistic studies have identified inflammation, pneumolysin, platelet activation, and thrombus formation at the center of cardiovascular disease progression. SUMMARY In this review, we will detail current knowledge of the mechanistic interaction between pneumonia and development of cardiovascular disease as well as discuss the current and potential drug therapy targets.
Collapse
|
18
|
Treatment With Acetylsalicylic Acid Reverses Endotoxin Tolerance in Humans In Vivo: A Randomized Placebo-Controlled Study. Crit Care Med 2020; 47:508-516. [PMID: 30585832 PMCID: PMC6426341 DOI: 10.1097/ccm.0000000000003630] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To investigate immunostimulatory effects of acetylsalicylic acid during experimental human endotoxemia and in sepsis patients. DESIGN Double-blind, randomized, placebo-controlled study in healthy volunteers and ex vivo stimulation experiments using monocytes of septic patients. SETTING Intensive care research unit of an university hospital. SUBJECTS Thirty healthy male volunteers and four sepsis patients. INTERVENTIONS Healthy volunteers were challenged IV with endotoxin twice, at a 1-week interval, with each challenge consisting of a bolus of 1 ng/kg followed by continuous administration of 1 ng/kg/hr during 3 hours. Volunteers were randomized to acetylsalicylic acid prophylaxis (80 mg acetylsalicylic acid daily for a 14-d period, starting 7 d before the first endotoxin challenge), acetylsalicylic acid treatment (80 mg acetylsalicylic acid daily for the 7-d period in-between both endotoxin challenges), or the control group (receiving placebo). Furthermore, monocytes of sepsis patients were incubated with acetylsalicylic acid preexposed platelets and were subsequently stimulated with endotoxin. MEASUREMENTS AND MAIN RESULTS Acetylsalicylic acid prophylaxis enhanced plasma tumor necrosis factor-α concentrations upon the first endotoxin challenge by 50% compared with the control group (p = 0.02) but did not modulate cytokine responses during the second endotoxin challenge. In contrast, acetylsalicylic acid treatment resulted in enhanced plasma levels of tumor necrosis factor-α (+53%; p = 0.02), interleukin-6 (+91%; p = 0.03), and interleukin-8 (+42%; p = 0.02) upon the second challenge, whereas plasma levels of the key antiinflammatory cytokine interleukin-10 were attenuated (-40%; p = 0.003). This proinflammatory phenotype in the acetylsalicylic acid treatment group was accompanied by a decrease in urinary prostaglandin E metabolite levels (-27% ± 7%; p = 0.01). Ex vivo exposure of platelets to acetylsalicylic acid increased production of tumor necrosis factor-α (+66%) and decreased production of interleukin-10 (-23%) by monocytes of sepsis patients. CONCLUSIONS Treatment, but not prophylaxis, with low-dose acetylsalicylic acid, partially reverses endotoxin tolerance in humans in vivo by shifting response toward a proinflammatory phenotype. This acetylsalicylic acid-induced proinflammatory shift was also observed in septic monocytes, signifying that patients suffering from sepsis-induced immunoparalysis might benefit from initiating acetylsalicylic acid treatment.
Collapse
|
19
|
Tiwari NR, Chaudhari KS, Sharma R, Haas KP, Sharma VR. Antiplatelet Agents in Sepsis-Putting it all together: A Call to Action. Indian J Crit Care Med 2020; 24:483-484. [PMID: 32863646 PMCID: PMC7435098 DOI: 10.5005/jp-journals-10071-23450] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
How to cite this article: Tiwari NR, Chaudhari KS, Sharma R, Haas KP, Sharma VR. Antiplatelet Agents in Sepsis-Putting it all together: A Call to Action. Indian J Crit Care Med 2020;24(6):483-484.
Collapse
Affiliation(s)
- Nishant R Tiwari
- Department of Internal Medicine, Byramjee Jeejeebhoy Government Medical College, Pune, Maharashtra, India
| | - Kaustubh S Chaudhari
- Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma, USA
| | - Richa Sharma
- Department of Ophthalmology, Byramjee Jeejeebhoy Government Medical College, Pune, Maharashtra, India
| | - Kevin P Haas
- Department of Pulmonary and Critical Care Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Vivek R Sharma
- Department of Hematology and Oncology, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
20
|
Carestia A, Davis RP, Davis L, Jenne CN. Inhibition of immunothrombosis does not affect pathogen capture and does not promote bacterial dissemination in a mouse model of sepsis. Platelets 2019; 31:925-931. [PMID: 31851856 DOI: 10.1080/09537104.2019.1704711] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
After infection, neutrophils release neutrophil extracellular traps (NETs), decondensed DNA fibers decorated with both nuclear proteins and proteins derived from intracellular granules. These structures have a fundamental role in the development of immunothrombosis; a physiological process mediated by immune cells and molecules from the coagulation system that facilitates the recognition, containment, and destruction of pathogens. Although NETs and immunothrombi are widely hypothesized to be key host defense responses responsible for limiting bacterial dissemination, their actual role in this process has not been formally assessed within the context of a bloodstream infection. Mice were first treated with LPS to generate inflammation (NETs and immunothrombi) and then bacteria dissemination was analyzed by intravital microscopy and colony-forming units (CFU) assay. Blocking NETs or coagulation by the administration of DNase or Argatroban (thrombin inhibitor), respectively, did not modify the percentage of bacteria capture by Kupffer cells, neutrophils or platelets. Moreover, both inhibitors reduced the number of bacteria in the spleen, without modifying CFUs in the liver or lung. In conclusion, we demonstrate that immunothrombi are not necessary to limit the dissemination of bloodstream bacterial infections.
Collapse
Affiliation(s)
- Agostina Carestia
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary , Calgary, Canada
| | - Rachelle P Davis
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary , Calgary, Canada
| | - Lauren Davis
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham , Birmingham, UK
| | - Craig N Jenne
- Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary , Calgary, Canada
| |
Collapse
|
21
|
Davidson JA, Warren-Gash C. Cardiovascular complications of acute respiratory infections: current research and future directions. Expert Rev Anti Infect Ther 2019; 17:939-942. [PMID: 31684779 DOI: 10.1080/14787210.2019.1689817] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Jennifer A Davidson
- Department of Non-communicable Diseases Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK
| | - Charlotte Warren-Gash
- Department of Non-communicable Diseases Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW Platelets are specialized effector cells that rapidly respond to sites of vascular injury. However, emerging data demonstrate that platelets possess diverse functions that also mediate inflammatory responses and neurological diseases. These functions are relevant to disease processes prevalent among older adults and likely influence susceptibility to thrombotic and inflammatory disorders. RECENT FINDINGS Platelet counts decrease in aged individuals whereas platelet reactivity increases. The platelet transcriptome is altered in aged individuals resulting in altered platelet function and exaggerated inflammation. Platelet signaling to monocytes in aging results in significantly more cytokines because of increased platelet-derived granzyme A. Platelet activation in aging appears to be driven, in part, because of increased reactive oxygen species and activation of the mammalian target of rapamycin pathway. Increased platelet hyperactivity in diseases is associated with aging, such cardiovascular disease and sepsis, exaggerate inflammation and thrombosis. Noncanonical functions of platelets influence the development of neurological diseases including Alzheimer's disease. SUMMARY Although there have been advances dissecting the molecular mechanisms regarding aging-related changes in platelets, many knowledge gaps still remain. Studies filling these gaps are likely to identify new mechanisms driving aging-related changes in platelet gene expression and function, and contributing to injurious thrombo-inflammation in older adults.
Collapse
Affiliation(s)
- Emilie Montenont
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, 84112
| | - Matthew T. Rondina
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, 84112
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, 84132
- George E. Wahlen VAMC Department of Internal Medicine and GRECC, Salt Lake City, Utah, 84148
| | - Robert A. Campbell
- University of Utah Molecular Medicine Program, Salt Lake City, Utah, 84112
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, 84132
| |
Collapse
|
23
|
Vardon-Bounes F, Ruiz S, Gratacap MP, Garcia C, Payrastre B, Minville V. Platelets Are Critical Key Players in Sepsis. Int J Mol Sci 2019; 20:ijms20143494. [PMID: 31315248 PMCID: PMC6679237 DOI: 10.3390/ijms20143494] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 07/07/2019] [Accepted: 07/08/2019] [Indexed: 01/13/2023] Open
Abstract
Host defense against infection is based on two crucial mechanisms: the inflammatory response and the activation of coagulation. Platelets are involved in both hemostasis and immune response. These mechanisms work together in a complex and synchronous manner making the contribution of platelets of major importance in sepsis. This is a summary of the pathophysiology of sepsis-induced thrombocytopenia, microvascular consequences, platelet-endothelial cells and platelet–pathogens interactions. The critical role of platelets during sepsis and the therapeutic implications are also reviewed.
Collapse
Affiliation(s)
- Fanny Vardon-Bounes
- Anesthesiology and Critical Care Unit, Toulouse University Hospital, 31059 Toulouse, France.
- INSERM I2MC (Institut des Maladies Cardiovasculaires et Métaboliques) UMR 1048, Toulouse University Hospital, 31059 Toulouse, France.
| | - Stéphanie Ruiz
- Anesthesiology and Critical Care Unit, Toulouse University Hospital, 31059 Toulouse, France
| | - Marie-Pierre Gratacap
- INSERM I2MC (Institut des Maladies Cardiovasculaires et Métaboliques) UMR 1048, Toulouse University Hospital, 31059 Toulouse, France
| | - Cédric Garcia
- Hematology Laboratory, Toulouse University Hospital, 31059 Toulouse, France
| | - Bernard Payrastre
- INSERM I2MC (Institut des Maladies Cardiovasculaires et Métaboliques) UMR 1048, Toulouse University Hospital, 31059 Toulouse, France
- Hematology Laboratory, Toulouse University Hospital, 31059 Toulouse, France
| | - Vincent Minville
- Anesthesiology and Critical Care Unit, Toulouse University Hospital, 31059 Toulouse, France
- INSERM I2MC (Institut des Maladies Cardiovasculaires et Métaboliques) UMR 1048, Toulouse University Hospital, 31059 Toulouse, France
| |
Collapse
|
24
|
Ouyang Y, Wang Y, Liu B, Ma X, Ding R. Effects of antiplatelet therapy on the mortality rate of patients with sepsis: A meta-analysis. J Crit Care 2018; 50:162-168. [PMID: 30551047 DOI: 10.1016/j.jcrc.2018.12.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 11/30/2018] [Accepted: 12/04/2018] [Indexed: 01/11/2023]
Abstract
PURPOSE Abnormal platelet activation plays an important role in the development of sepsis. The effect of antiplatelet drugs on the outcome of patients with sepsis remains unclear. This meta-analysis aimed to determine the effect of antiplatelet drugs on the prognosis of patients with sepsis. MATERIALS AND METHODS PubMed, Cochrane Library, CBM, and Embase were searched for all related articles published from inception to April 2018. The primary end point was mortality. Adjusted data were used and statistically analysed. RESULTS Ten cohort studies were included. The total number of patients with sepsis was 689,897. Data showed that the use of antiplatelet drugs could effectively reduce the mortality of patients with sepsis (odds ratio (OR) = 0.82, 95% CI: 0.81-0.83, p < 0.05). Seven studies used aspirin for antiplatelet therapy, and subgroup analysis showed that aspirin effectively reduced ICU or hospital mortality in patients with sepsis (OR = 0.60, 95% CI: 0.53-0.68, p < 0.05). A subgroup analysis on the timing of anti-platelet drug administration showed that antiplatelet drugs can reduce mortality when administered either before (OR = 0.78, 95% CI: 0.77-0.80) or after sepsis (OR = 0.59, 95% CI: 0.52-0.67). CONCLUSIONS Antiplatelet drugs, particularly aspirin, could be used to effectively reduce mortality in patients with sepsis.
Collapse
Affiliation(s)
- Yaqi Ouyang
- Department of Intensive Care Unit, Xiehe Hospital affiliated to Tongji Medical College, Huazhong University of Science and Technology,Jiefang Avenue 1277, Wuhan 430000, , Hubei Province, PR China
| | - Yuhui Wang
- Department of Intensive Care Unit, The First Hospital of China Medical University, Nanjing Bei Street 155, Shenyang 110001, Liaoning Province, PR China
| | - Baoyan Liu
- Department of Intensive Care Unit, The First Hospital of China Medical University, Nanjing Bei Street 155, Shenyang 110001, Liaoning Province, PR China
| | - Xiaochun Ma
- Department of Intensive Care Unit, The First Hospital of China Medical University, Nanjing Bei Street 155, Shenyang 110001, Liaoning Province, PR China.
| | - Renyu Ding
- Department of Intensive Care Unit, The First Hospital of China Medical University, Nanjing Bei Street 155, Shenyang 110001, Liaoning Province, PR China.
| |
Collapse
|
25
|
Kim TJ, Lee JS, Kang MK, Nam KW, Lee CH, Mo H, Jeong HY, Yoon BW, Ko SB. Clopidogrel may decrease the risk of post-stroke infection after ischaemic stroke. Eur J Neurol 2018; 26:261-267. [PMID: 30168901 DOI: 10.1111/ene.13801] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 08/28/2018] [Indexed: 01/09/2023]
Abstract
BACKGROUND AND PURPOSE The P2Y12 receptor, a well-known factor in the platelet activation pathway, plays a role in thrombosis as well as systemic inflammation. Clopidogrel, a prototype P2Y12 receptor antagonist, reportedly decreases inflammation and systemic infection. The aim of this study was to evaluate whether clopidogrel use decreases the risk of post-stroke infection following ischaemic stroke. METHODS A total of 1643 patients with acute ischaemic stroke (within 7 days after onset) were included for analysis between March 2010 and December 2015. Patients were categorized into two groups (clopidogrel users versus clopidogrel non-users), and clinical characteristics and risks of post-stroke infection were compared between the two groups. The inverse probability of treatment weighting using propensity scores for baseline imbalance adjustments was applied. RESULTS Of the included patients (mean age 67.7 years; men 60.6%), 670 (40.8%) patients were clopidogrel users and 164 (10.0%) patients had post-stroke infection. The proportion of patients with post-stroke infection was significantly lower in clopidogrel users compared to clopidogrel non-users (6.7% vs. 12.2%, P ≤ 0.001). Moreover, clopidogrel users were less likely to be admitted to the intensive care unit (13.3% vs. 35.3%, P = 0.006). A multivariate analysis with inverse probability of treatment weighting revealed that clopidogrel users exhibited a lower risk of post-stroke infection (odds ratio 0.56, 95% confidence interval 0.42-0.75) and intensive care unit admission (odds ratio 0.34, 95% confidence interval 0.22-0.53). CONCLUSIONS The study suggested that clopidogrel users exhibit a lower risk of infection and develop less severe infections after ischaemic stroke. Further prospective studies are needed.
Collapse
Affiliation(s)
- T J Kim
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
| | - J S Lee
- Clinical Research Center, Asan Medical Center, Seoul, Korea
| | - M-K Kang
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
| | - K-W Nam
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
| | - C-H Lee
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
| | - H Mo
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
| | - H-Y Jeong
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
| | - B-W Yoon
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
| | - S-B Ko
- Department of Neurology, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
26
|
Sexton TR, Zhang G, Macaulay TE, Callahan LA, Charnigo R, Vsevolozhskaya OA, Li Z, Smyth S. Ticagrelor Reduces Thromboinflammatory Markers in Patients With Pneumonia. JACC Basic Transl Sci 2018; 3:435-449. [PMID: 30175268 PMCID: PMC6115703 DOI: 10.1016/j.jacbts.2018.05.005] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 05/03/2018] [Indexed: 01/16/2023]
Abstract
As expected, ticagrelor reduced ex-vivo ADP-induced aggregation in patients with pneumonia compared with placebo. Ticagrelor reduced platelet–leukocyte interactions as well as plasma interleukin-6 within 24 h in patients with pneumonia compared with placebo. Ticagrelor acutely altered NETosis biomarkers, whereas placebo had no effect. Ticagrelor improved lung function and reduced need for supplemental oxygen in patients with pneumonia compared with placebo.
Despite treatment advances for sepsis and pneumonia, significant improvements in outcome have not been realized. Antiplatelet therapy may improve outcome in pneumonia and sepsis. In this study, the authors show that ticagrelor reduced leukocytes with attached platelets as well as the inflammatory biomarker interleukin (IL)-6. Pneumonia patients receiving ticagrelor required less supplemental oxygen and lung function tests trended toward improvement. Disruption of the P2Y12 receptor in a murine model protected against inflammatory response, lung permeability, and mortality. Results indicate a mechanistic link between platelets, leukocytes, and lung injury in settings of pneumonia and sepsis, and suggest possible therapeutic approaches to reduce complications.(Targeting Platelet-Leukocyte Aggregates in Pneumonia With Ticagrelor [XANTHIPPE]; NCT01883869)
Collapse
Key Words
- ADP, adenosine diphosphate
- CAP, community-acquired pneumonia
- CI, confidence interval
- COPD, chronic obstructive pulmonary disease
- ELISA, enzyme-linked immunosorbent assay
- FEV-1, forced expiratory volume in 1 s
- HAP, hospital-acquired pneumonia
- IL, interleukin
- IQR, interquartile range
- Kfc, capillary filtration coefficient
- LPS, lipopolysaccharide
- LTA, light transmission aggregometry
- MPO, myeloperoxidase
- MVV, maximum ventilation velocity
- NE, neutrophil elastase
- NET, neutrophil extracellular trap
- OR, odds ratio
- PRP, platelet-rich plasma
- TNF, tumor necrosis factor
- TRAP, thrombin receptor activating peptide
- WT, wild-type
- dsDNA, doubled-stranded DNA
- inflammation
- leukocytes
- platelets
- pneumonia
- sepsis
Collapse
Affiliation(s)
- Travis R Sexton
- Gill Heart and Vascular Institute, University of Kentucky, Lexington, Kentucky
| | - Guoying Zhang
- Gill Heart and Vascular Institute, University of Kentucky, Lexington, Kentucky
| | - Tracy E Macaulay
- Gill Heart and Vascular Institute, University of Kentucky, Lexington, Kentucky
| | - Leigh A Callahan
- Pulmonary, Critical Care & Sleep Medicine, University of Kentucky, Lexington, Kentucky
| | - Richard Charnigo
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, Kentucky
| | - Olga A Vsevolozhskaya
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, Kentucky
| | - Zhenyu Li
- Gill Heart and Vascular Institute, University of Kentucky, Lexington, Kentucky
| | - Susan Smyth
- Gill Heart and Vascular Institute, University of Kentucky, Lexington, Kentucky.,Lexington VA Medical Center, Lexington, Kentucky
| |
Collapse
|
27
|
Wang Y, Ouyang Y, Liu B, Ma X, Ding R. Platelet activation and antiplatelet therapy in sepsis: A narrative review. Thromb Res 2018; 166:28-36. [DOI: 10.1016/j.thromres.2018.04.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/03/2018] [Accepted: 04/06/2018] [Indexed: 12/31/2022]
|
28
|
The preventive effect of antiplatelet therapy in acute respiratory distress syndrome: a meta-analysis. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2018. [PMID: 29519254 PMCID: PMC5844104 DOI: 10.1186/s13054-018-1988-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Acute respiratory distress syndrome (ARDS) is a life-threatening condition with high mortality that imposes a serious medical burden. Antiplatelet therapy is a potential strategy for preventing ARDS in patients with a high risk of developing this condition. A meta-analysis was performed to investigate whether antiplatelet therapy could reduce the incidence of newly developed ARDS and its associated mortality in high-risk patients. Methods The Cochrane Central Register of Controlled Trials (CENTRAL), PubMed, Embase, Medline, and the Web of Science were searched for published studies from inception to 26 October 2017. We included randomized clinical trials, cohort studies and case-control studies investigating antiplatelet therapy in adult patients presenting to the hospital or ICU with a high risk for ARDS. Baseline patient characteristics, interventions, controls and outcomes were extracted. Our primary outcome was the incidence of newly developed ARDS in high-risk patients. Secondary outcomes were hospital and ICU mortality. A random-effects or fixed-effects model was used for quantitative synthesis. Results We identified nine eligible studies including 7660 high-risk patients who received antiplatelet therapy. Based on seven observational studies, antiplatelet therapy was associated with a decreased incidence of ARDS (odds ratio (OR) 0.68, 95% confidence interval (CI) 0.52–0.88; I2 = 68.4%, p = 0.004). In two randomized studies, no significant difference was found in newly developed ARDS between the antiplatelet groups and placebo groups (OR 1.32, 95% CI 0.72–2.42; I2 = 0.0%, p = 0.329). Antiplatelet therapy did not reduce hospital mortality in randomized studies (OR 1.15, 95% CI 0.58–2.27; I2 = 0.0%; p = 0.440) or observational studies (OR 0.80, 95% CI 0.62–1.03; I2 = 31.9%, p = 0.221). Conclusions Antiplatelet therapy did not significantly decrease hospital mortality in high-risk patients. However, whether antiplatelet therapy is associated with a decreased incidence of ARDS in patients at a high risk of developing the condition remains unclear. Electronic supplementary material The online version of this article (10.1186/s13054-018-1988-y) contains supplementary material, which is available to authorized users.
Collapse
|
29
|
Abstract
Platelets play a vital role in normal hemostasis to stem blood loss at sites of vascular injury by tethering and adhering to sites of injury, recruiting other platelets and blood cells to the developing clot, releasing vasoactive small molecules and proteins, and assembling and activating plasma coagulation proteins in a tightly regulated temporal and spatial manner. In synchrony with specific end products of coagulation, primarily cross-linked fibrin, a stable thrombus quickly forms. Far beyond physiological hemostasis and pathological thrombosis, emerging evidence supports platelets playing a pivotal role in vascular homeostasis, inflammation, cellular repair, regeneration, and wide range of autocrine and paracrine functions. In essence, platelets play both structural and functional roles as reporters, messengers, and active transporters surveying the vasculature for cues of environmental or developmental stimuli and participating as first responders.1 In this review, we will provide a contemporary perspective of platelet physiology, including fundamental, translational, and clinical constructs that apply directly to human health and disease.
Collapse
Affiliation(s)
- Richard C Becker
- From the Heart, Lung and Vascular Institute, University of Cincinnati College of Medicine, OH (R.C.B.); and Gill Heart and Vascular Institute (T.S., S.S.S.) and Lexington VA Medical Center (T.S., S.S.S.), University of Kentucky School of Medicine.
| | - Travis Sexton
- From the Heart, Lung and Vascular Institute, University of Cincinnati College of Medicine, OH (R.C.B.); and Gill Heart and Vascular Institute (T.S., S.S.S.) and Lexington VA Medical Center (T.S., S.S.S.), University of Kentucky School of Medicine
| | - Susan S Smyth
- From the Heart, Lung and Vascular Institute, University of Cincinnati College of Medicine, OH (R.C.B.); and Gill Heart and Vascular Institute (T.S., S.S.S.) and Lexington VA Medical Center (T.S., S.S.S.), University of Kentucky School of Medicine
| |
Collapse
|
30
|
Dewitte A, Lepreux S, Villeneuve J, Rigothier C, Combe C, Ouattara A, Ripoche J. Blood platelets and sepsis pathophysiology: A new therapeutic prospect in critically [corrected] ill patients? Ann Intensive Care 2017; 7:115. [PMID: 29192366 PMCID: PMC5709271 DOI: 10.1186/s13613-017-0337-7] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 11/12/2017] [Indexed: 02/06/2023] Open
Abstract
Beyond haemostasis, platelets have emerged as versatile effectors of the immune response. The contribution of platelets in inflammation, tissue integrity and defence against infections has considerably widened the spectrum of their role in health and disease. Here, we propose a narrative review that first describes these new platelet attributes. We then examine their relevance to microcirculatory alterations in multi-organ dysfunction, a major sepsis complication. Rapid progresses that are made on the knowledge of novel platelet functions should improve the understanding of thrombocytopenia, a common condition and a predictor of adverse outcome in sepsis, and may provide potential avenues for management and therapy.
Collapse
Affiliation(s)
- Antoine Dewitte
- INSERM U1026, BioTis, Univ. Bordeaux, 33000, Bordeaux, France. .,Department of Anaesthesia and Critical Care II, Magellan Medico-Surgical Center, CHU Bordeaux, 33000, Bordeaux, France.
| | - Sébastien Lepreux
- INSERM U1026, BioTis, Univ. Bordeaux, 33000, Bordeaux, France.,Department of Pathology, CHU Bordeaux, 33000, Bordeaux, France
| | - Julien Villeneuve
- Cell and Developmental Biology Department, Centre for Genomic Regulation, The Barcelona Institute for Science and Technology, 08003, Barcelona, Spain
| | - Claire Rigothier
- INSERM U1026, BioTis, Univ. Bordeaux, 33000, Bordeaux, France.,Department of Nephrology, Transplantation and Haemodialysis, CHU Bordeaux, 33000, Bordeaux, France
| | - Christian Combe
- INSERM U1026, BioTis, Univ. Bordeaux, 33000, Bordeaux, France.,Department of Nephrology, Transplantation and Haemodialysis, CHU Bordeaux, 33000, Bordeaux, France
| | - Alexandre Ouattara
- Department of Anaesthesia and Critical Care II, Magellan Medico-Surgical Center, CHU Bordeaux, 33000, Bordeaux, France.,INSERM U1034, Biology of Cardiovascular Diseases, Univ. Bordeaux, 33600, Pessac, France
| | - Jean Ripoche
- INSERM U1026, BioTis, Univ. Bordeaux, 33000, Bordeaux, France
| |
Collapse
|
31
|
Quantifying the Effects of Prior Acetyl-Salicylic Acid on Sepsis-Related Deaths: An Individual Patient Data Meta-Analysis Using Propensity Matching. Crit Care Med 2017; 45:1871-1879. [PMID: 28799949 DOI: 10.1097/ccm.0000000000002654] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE The primary objective was to conduct a meta-analysis on published observational cohort data describing the association between acetyl-salicylic acid (aspirin) use prior to the onset of sepsis and mortality in hospitalized patients. STUDY SELECTION Studies that reported mortality in patients on aspirin with sepsis with a comparison group of patients with sepsis not on prior aspirin therapy were included. DATA SOURCES Fifteen studies described hospital-based cohorts (n = 17,065), whereas one was a large insurance-based database (n = 683,421). Individual-level patient data were incorporated from all selected studies. DATA EXTRACTION Propensity analyses with 1:1 propensity score matching at the study level were performed, using the most consistently available covariates judged to be associated with aspirin. Meta-analyses were performed to estimate the pooled average treatment effect of aspirin on sepsis-related mortality. DATA SYNTHESIS Use of aspirin was associated with a 7% (95% CI, 2-12%; p = 0.005) reduction in the risk of death as shown by meta-analysis with considerable statistical heterogeneity (I = 61.6%). CONCLUSIONS These results are consistent with effects ranging from a 2% to 12% reduction in mortality risk in patients taking aspirin prior to sepsis onset. This association anticipates results of definitive studies of the use of low-dose aspirin as a strategy for reduction of deaths in patients with sepsis.
Collapse
|
32
|
Greco E, Lupia E, Bosco O, Vizio B, Montrucchio G. Platelets and Multi-Organ Failure in Sepsis. Int J Mol Sci 2017; 18:ijms18102200. [PMID: 29053592 PMCID: PMC5666881 DOI: 10.3390/ijms18102200] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/13/2017] [Accepted: 10/17/2017] [Indexed: 12/11/2022] Open
Abstract
Platelets have received increasing attention for their role in the pathophysiology of infectious disease, inflammation, and immunity. In sepsis, a low platelet count is a well-known biomarker for disease severity and more recently authors have focused their attention on the active role of platelets in the pathogenesis of multi-organ failure. Septic shock is characterised by a dysregulated inflammatory response, which can impair the microcirculation and lead to organ injury. Being at the crossroads between the immune system, clotting cascade, and endothelial cells, platelets seem to be an appealing central mediator and possible therapeutic target in sepsis. This review focuses on the pathogenic role of platelets in septic organ dysfunction in humans and animal models.
Collapse
Affiliation(s)
- Elisabetta Greco
- Department of Medical Science, University of Turin, 10126 Turin, Italy.
| | - Enrico Lupia
- Department of Medical Science, University of Turin, 10126 Turin, Italy.
| | - Ornella Bosco
- Department of Medical Science, University of Turin, 10126 Turin, Italy.
| | - Barbara Vizio
- Department of Medical Science, University of Turin, 10126 Turin, Italy.
| | | |
Collapse
|
33
|
Layios N, Delierneux C, Hego A, Huart J, Gosset C, Lecut C, Maes N, Geurts P, Joly A, Lancellotti P, Albert A, Damas P, Gothot A, Oury C. Sepsis prediction in critically ill patients by platelet activation markers on ICU admission: a prospective pilot study. Intensive Care Med Exp 2017; 5:32. [PMID: 28699088 PMCID: PMC5505890 DOI: 10.1186/s40635-017-0145-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 07/03/2017] [Indexed: 01/17/2023] Open
Abstract
Background Platelets have been involved in both immune surveillance and host defense against severe infection. To date, whether platelet phenotype or other hemostasis components could be associated with predisposition to sepsis in critical illness remains unknown. The aim of this work was to identify platelet markers that could predict sepsis occurrence in critically ill injured patients. Methods This single-center, prospective, observational, 7-month study was based on a cohort of 99 non-infected adult patients admitted to ICUs for elective cardiac surgery, trauma, acute brain injury, and post-operative prolonged ventilation and followed up during ICU stay. Clinical characteristics and severity score (SOFA) were recorded on admission. Platelet activation markers, including fibrinogen binding to platelets, platelet membrane P-selectin expression, plasma soluble CD40L, and platelet-leukocytes aggregates were assayed by flow cytometry at admission and 48 h later, and then at the time of sepsis diagnosis (Sepsis-3 criteria) and 7 days later for sepsis patients. Hospitalization data and outcomes were also recorded. Methods Of the 99 patients, 19 developed sepsis after a median time of 5 days. These patients had a higher SOFA score at admission; levels of fibrinogen binding to platelets (platelet-Fg) and of D-dimers were also significantly increased compared to the other patients. Levels 48 h after ICU admission no longer differed between the two patient groups. Platelet-Fg % was an independent predictor of sepsis (P = 0.0031). By ROC curve analysis, cutoff point for Platelet-Fg (AUC = 0.75) was 50%. In patients with a SOFA cutoff of 8, the risk of sepsis reached 87% when Platelet-Fg levels were above 50%. Patients with sepsis had longer ICU and hospital stays and higher death rate. Conclusions Platelet-bound fibrinogen levels assayed by flow cytometry within 24 h of ICU admission help identifying critically ill patients at risk of developing sepsis. Electronic supplementary material The online version of this article (doi:10.1186/s40635-017-0145-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nathalie Layios
- Department of General Intensive Care, University Hospital of Liège, Liège, Belgium.,Laboratory of Thrombosis and Hemostasis, GIGA-Cardiovascular Sciences, University of Liège, Department of Cardiology, University Hospital of Liège, Liège, Belgium
| | - Céline Delierneux
- Laboratory of Thrombosis and Hemostasis, GIGA-Cardiovascular Sciences, University of Liège, Department of Cardiology, University Hospital of Liège, Liège, Belgium
| | - Alexandre Hego
- Laboratory of Thrombosis and Hemostasis, GIGA-Cardiovascular Sciences, University of Liège, Department of Cardiology, University Hospital of Liège, Liège, Belgium
| | - Justine Huart
- Laboratory of Thrombosis and Hemostasis, GIGA-Cardiovascular Sciences, University of Liège, Department of Cardiology, University Hospital of Liège, Liège, Belgium
| | - Christian Gosset
- Laboratory of Hematology, University Hospital of Liège, Liège, Belgium
| | - Christelle Lecut
- Laboratory of Hematology, University Hospital of Liège, Liège, Belgium
| | - Nathalie Maes
- Department of Biostatistics and Medico-Economic Information, University Hospital of Liège, Liège, Belgium
| | - Pierre Geurts
- Systems and Modeling, Department of Electrical Engineering and Computer Science, University of Liège, Liège, Belgium
| | - Arnaud Joly
- Systems and Modeling, Department of Electrical Engineering and Computer Science, University of Liège, Liège, Belgium
| | - Patrizio Lancellotti
- Laboratory of Thrombosis and Hemostasis, GIGA-Cardiovascular Sciences, University of Liège, Department of Cardiology, University Hospital of Liège, Liège, Belgium.,Gruppo Villa Maria Care and Research, Anthea Hospital, Bari, Italy
| | - Adelin Albert
- Department of Biostatistics and Medico-Economic Information, University Hospital of Liège, Liège, Belgium
| | - Pierre Damas
- Department of General Intensive Care, University Hospital of Liège, Liège, Belgium
| | - André Gothot
- Laboratory of Hematology, University Hospital of Liège, Liège, Belgium
| | - Cécile Oury
- Laboratory of Thrombosis and Hemostasis, GIGA-Cardiovascular Sciences, University of Liège, Department of Cardiology, University Hospital of Liège, Liège, Belgium.
| |
Collapse
|
34
|
Kiers D, van der Heijden WA, van Ede L, Gerretsen J, de Mast Q, van der Ven AJ, El Messaoudi S, Rongen GA, Gomes M, Kox M, Pickkers P, Riksen NP. A randomised trial on the effect of anti-platelet therapy on the systemic inflammatory response in human endotoxaemia. Thromb Haemost 2017; 117:1798-1807. [PMID: 28692111 DOI: 10.1160/th16-10-0799] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 05/25/2017] [Indexed: 01/09/2023]
Abstract
The use of acetylsalicylic acid (ASA) is associated with improved outcome in patients with sepsis, and P2Y12 inhibitors have been suggested to also have immunomodulatory effects. Therefore, we evaluated the effects of clinically relevant combinations of antiplatelet therapy on the immune response in experimental endotoxaemia in humans in vivo. Forty healthy subjects were randomised to seven days of placebo, placebo with ASA, ticagrelor and ASA, or clopidogrel and ASA treatment. Systemic inflammation was elicited at day seven by intravenous administration of Escherichia coli endotoxin. ASA treatment profoundly augmented the plasma concentration of pro-inflammatory cytokines, but did not affect anti-inflammatory cytokines. Addition of either P2Y12 antagonist to ASA did not affect any of the circulating cytokines, except for an attenuation of the ASA-induced increase in TNFα by ticagrelor. Systemic inflammation increased plasma adenosine, without differences between groups, and although P2Y12 inhibition impaired platelet reactivity, there was no correlation with cytokine responses.
Collapse
Affiliation(s)
- Dorien Kiers
- Prof. N. P. Riksen, Dept. of Internal Medicine 463, Radboud University Medical Center, PO Box 9101, 6500HB Nijmegen, The Netherlands, Tel.: +31 24 3618819, Fax: +31 24 3616519, E-mail:
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Mohananey D, Sethi J, Villablanca PA, Ali MS, Kumar R, Baruah A, Bhatia N, Agrawal S, Hussain Z, Shamoun FE, Augoustides JT, Ramakrishna H. Effect of antiplatelet therapy on mortality and acute lung injury in critically ill patients: A systematic review and meta-analysis. Ann Card Anaesth 2017; 19:626-637. [PMID: 27716693 PMCID: PMC5070322 DOI: 10.4103/0971-9784.191576] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Aim: Platelet function is intricately linked to the pathophysiology of critical Illness, and some studies have shown that antiplatelet therapy (APT) may decrease mortality and incidence of acute respiratory distress syndrome (ARDS) in these patients. Our objective was to understand the efficacy of APT by conducting a meta-analysis. Materials and Methods: We conducted a meta-analysis using PubMed, Central, Embase, The Cochrane Central Register, the ClinicalTrials.gov Website, and Google Scholar. Studies were included if they investigated critically ill patients receiving antiplatelet therapy and mentioned the outcomes being studied (mortality, duration of hospitalization, ARDS, and need for mechanical ventilation). Results: We found that there was a significant reduction in all-cause mortality in patients on APT compared to control (odds ratio [OR]: 0.83; 95% confidence interval [CI]: 0.70–0.97). Both the incidence of acute lung injury/ARDS (OR: 0.67; 95% CI: 0.57–0.78) and need for mechanical ventilation (OR: 0.74; 95% CI: 0.60–0.91) were lower in the antiplatelet group. No significant difference in duration of hospitalization was observed between the two groups (standardized mean difference: −0.02; 95% CI: −0.11–0.07). Conclusion: Our meta-analysis suggests that critically ill patients who are on APT have an improved survival, decreased incidence of ARDS, and decreased need for mechanical ventilation.
Collapse
Affiliation(s)
- Divyanshu Mohananey
- Department of Internal Medicine, John H Stroger, Jr. Hospital of Cook County, Chicago, IL 60612, USA
| | - Jaskaran Sethi
- Department of Internal Medicine, John H Stroger, Jr. Hospital of Cook County, Chicago, IL 60612, USA
| | - Pedro A Villablanca
- Division of Cardiovascular Diseases, Albert Einstein College of Medicine, Montefiore Medical Center, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Muhammad S Ali
- Department of Internal Medicine, John H Stroger, Jr. Hospital of Cook County, Chicago, IL 60612, USA
| | - Rohit Kumar
- Department of Internal Medicine, John H Stroger, Jr. Hospital of Cook County, Chicago, IL 60612, USA
| | - Anushka Baruah
- Department of Internal Medicine, John H Stroger, Jr. Hospital of Cook County, Chicago, IL 60612, USA
| | - Nirmanmoh Bhatia
- Division of Cardiovascular Medicine, Vanderbilt Heart and Vascular Institute, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, USA
| | - Sahil Agrawal
- Department of Cardiovascular Medicine, St. Lukes University Health Network, Bethlehem, PA 18015, USA
| | - Zeeshan Hussain
- Department of Internal Medicine, University of Louisville, Louisville, KY 40292, USA
| | - Fadi E Shamoun
- Division of Cardiovascular Diseases, 13400 East Mayo Boulevard, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - John T Augoustides
- Department of Anesthesiology, Hospital of the University of Pennsylvania, 6 Dulles, Philadelphia, PA 19104, USA
| | - Harish Ramakrishna
- Department of Anesthesiology, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, USA
| |
Collapse
|
36
|
Anderson R, Feldman C. Pneumolysin as a potential therapeutic target in severe pneumococcal disease. J Infect 2017; 74:527-544. [PMID: 28322888 DOI: 10.1016/j.jinf.2017.03.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 03/09/2017] [Accepted: 03/11/2017] [Indexed: 12/13/2022]
Abstract
Acute pulmonary and cardiac injury remain significant causes of morbidity and mortality in those afflicted with severe pneumococcal disease, with the risk for early mortality often persisting several years beyond clinical recovery. Although remaining to be firmly established in the clinical setting, a considerable body of evidence, mostly derived from murine models of experimental infection, has implicated the pneumococcal, cholesterol-binding, pore-forming toxin, pneumolysin (Ply), in the pathogenesis of lung and myocardial dysfunction. Topics covered in this review include the burden of pneumococcal disease, risk factors, virulence determinants of the pneumococcus, complications of severe disease, antibiotic and adjuvant therapies, as well as the structure of Ply and the role of the toxin in disease pathogenesis. Given the increasing recognition of the clinical potential of Ply-neutralisation strategies, the remaining sections of the review are focused on updates of the types, benefits and limitations of currently available therapies which may attenuate, directly and/or indirectly, the injurious actions of Ply. These include recently described experimental therapies such as various phytochemicals and lipids, and a second group of more conventional agents the members of which remain the subject of ongoing clinical evaluation. This latter group, which is covered more extensively, encompasses macrolides, statins, corticosteroids, and platelet-targeted therapies, particularly aspirin.
Collapse
Affiliation(s)
- Ronald Anderson
- Department of Immunology and Institute of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.
| | - Charles Feldman
- Division of Pulmonology, Department of Internal Medicine, Charlotte Maxeke Johannesburg Academic Hospital and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
37
|
Eisen DP, Moore EM, Leder K, Lockery J, McBryde ES, McNeil JJ, Pilcher D, Wolfe R, Woods RL. AspiriN To Inhibit SEPSIS (ANTISEPSIS) randomised controlled trial protocol. BMJ Open 2017; 7:e013636. [PMID: 28110287 PMCID: PMC5253551 DOI: 10.1136/bmjopen-2016-013636] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Sepsis is a leading global cause of morbidity and mortality, and is more common at the extremes of age. Moreover, the cost of in-hospital care for elderly patients with sepsis is significant. There are indications from experimental and observational studies that aspirin may reduce inflammation associated with infection. This paper describes the rationale and design of the AspiriN To Inhibit SEPSIS (ANTISEPSIS) trial, a substudy of ASPirin in Reducing Events in the Elderly (ASPREE). ANTISEPSIS primarily aims to determine whether low-dose aspirin reduces sepsis-related deaths in older people. Additionally, it will assess whether low-dose aspirin reduces sepsis-related hospitalisations and sepsis-related Intensive Care Unit (ICU) admissions. METHODS AND ANALYSIS ASPREE is a double-blinded, randomised, placebo-controlled primary prevention trial that will determine whether daily low-dose aspirin extends disability-free longevity in 19 000 healthy older people recruited in Australia and the USA. The ANTISEPSIS substudy involves additional ASPREE trial data collection to assess the impact of daily low-dose aspirin on sepsis-related events in the 16 703 ASPREE participants aged 70 years and over, recruited in Australia. The intervention is a daily 100 mg dose of enteric-coated aspirin versus matching placebo, with 1:1 randomisation. The primary outcome for the ANTISEPSIS substudy is the incidence of sepsis-related death in eligible patients. The incidence of sepsis-related hospital and ICU admissions are secondary outcomes. ANTISEPSIS is to be conducted between 2012 and 2018. DISCUSSION This substudy will determine whether aspirin, an inexpensive and accessible therapy, safely reduces sepsis-related deaths and hospitalisations in older Australians. If shown to be the case, this would have profound effects on the health of older Australians. TRIAL REGISTRATION NUMBER Pre-results, ACTRN12613000349741.
Collapse
Affiliation(s)
- Damon P Eisen
- Townsville Hospital and Health Service, Townsville, Queensland, Australia
- Division of Tropical Health and Medicine, College of Medicine and Dentistry, James Cook University, Townsville, Queensland, Australia
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Elizabeth M Moore
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Karin Leder
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Victorian Infectious Diseases Service at the Peter Doherty Institute, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Jessica Lockery
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Emma S McBryde
- Division of Tropical Health and Medicine, Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Queensland, Australia
| | - John J McNeil
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - David Pilcher
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Department of Intensive Care, The Alfred Hospital, Melbourne, Victoria, Australia
- The Australian and New Zealand Intensive Care Society (ANZICS) Centre for Outcome and Resource Evaluation (CORE), Melbourne, Victoria, Australia
| | - Rory Wolfe
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Robyn L Woods
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
38
|
Tunjungputri RN, Peters E, van der Ven A, de Groot PG, de Mast Q, Pickkers P. Human recombinant alkaline phosphatase inhibits ex vivo platelet activation in humans. Thromb Haemost 2016; 116:1111-1121. [PMID: 27656709 DOI: 10.1160/th16-03-0206] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 09/07/2016] [Indexed: 12/15/2022]
Abstract
Sepsis-associated acute kidney injury (AKI) is associated with high morbidity and mortality. Excessive platelet activation contributes to AKI through the formation of microthrombi and amplification of systemic inflammation. Two phase II trials demonstrated that bovine-intestinal alkaline phosphatase (AP) improved renal function in critically ill patients with sepsis-associated AKI. In this study, we characterised the platelet-inhibiting effects of a human recombinant AP. Whole blood and platelet-rich plasma (PRP) of healthy volunteers (n=6) was pre-treated ex vivo with recAP, whereafter platelet reactivity to ADP, collagen-related peptide (CRP-XL) and Pam3CSK4 was determined by flow cytometry. RecAP (40 U/ml) reduced the platelet reactivity to ADP (inhibition with a median of 47 %, interquartile range 43-49 %; p<0.001) and tended to reduce platelet reactivity to CRP-XL (9 %, 2-25 %; p=0.08) in whole blood. The platelet-inhibiting effects of recAP were more pronounced in PRP both for ADP- (64 %, 54-68 %; p=0.002) and CRP-XL-stimulated samples (60 %, 46-71 %; p=0.002). RecAP rapidly converted ADP into adenosine, whereas antagonism of the A2A adenosine receptor partially reversed the platelet inhibitory effects of recAP. Platelets of septic shock patients (n=5) showed a 31% (22-34%; p=0.03) more pronounced reactivity compared to healthy volunteers, and this was completely reversed by recAP treatment. In conclusion, we demonstrate that recAP inhibits ex vivo human platelet activation through dephosphorylation of ADP and formation of adenosine as its turnover product. RecAP is able to reverse the platelet hyperreactivity present in septic shock patients. These effects may contribute to the beneficial effects of recAP as a new therapeutic candidate for sepsis-associated AKI.
Collapse
Affiliation(s)
| | | | | | | | | | - Peter Pickkers
- Peter Pickkers, Department of Intensive Care Medicine, Radboud university medical center, PO Box 9101, Internal Mailbag 710, 6500 HB Nijmegen, The Netherlands, Tel.: +31 24 36 15363, Fax: +31 24 36 68058, E-mail:
| |
Collapse
|
39
|
Feldman C, Anderson R. Prevalence, pathogenesis, therapy, and prevention of cardiovascular events in patients with community-acquired pneumonia. Pneumonia (Nathan) 2016; 8:11. [PMID: 28702290 PMCID: PMC5471702 DOI: 10.1186/s41479-016-0011-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 07/04/2016] [Indexed: 12/11/2022] Open
Abstract
It is now well recognised that cardiac events occur relatively commonly in patients with acute community-acquired pneumonia. While these events are more frequent in patients with underlying risk factors—such as those with underlying chronic cardiovascular and respiratory comorbidities, the elderly, and in nursing home residents—they also occur in patients with no underlying risks other than severe pneumonia. Recent research elucidating the underlying pathogenic mechanisms related to these cardiac events has indicated a probable role for platelet activation, which is possibly exacerbated by pneumolysin in the case of pneumococcal infections. This, in turn, has resulted in the identification of possible therapeutic strategies targeting platelet activation, as well as the cardio-toxic activity of pneumolysin. These issues represent the primary focus of the current review.
Collapse
Affiliation(s)
- Charles Feldman
- Division of Pulmonology, Department of Internal Medicine, Charlotte Maxeke Johannesburg Academic Hospital, Johannesburg, South Africa.,Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Department of Internal Medicine, University of the Witwatersrand Medical School, 7 York Road, Parktown, 2193 Johannesburg, South Africa
| | - Ronald Anderson
- Institute for Cellular and Molecular Medicine, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
40
|
Wang L, Li H, Gu X, Wang Z, Liu S, Chen L. Effect of Antiplatelet Therapy on Acute Respiratory Distress Syndrome and Mortality in Critically Ill Patients: A Meta-Analysis. PLoS One 2016; 11:e0154754. [PMID: 27182704 PMCID: PMC4868259 DOI: 10.1371/journal.pone.0154754] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Accepted: 04/19/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Antiplatelet agents are commonly used for cardiovascular diseases, but their pleiotropic effects in critically ill patients are controversial. We therefore performed a meta-analysis of cohort studies to investigate the effect of antiplatelet therapy in the critically ill. METHODS Nine cohort studies, retrieved from PubMed and Embase before November 2015, involving 14,612 critically ill patients and 4765 cases of antiplatelet users, were meta-analysed. The main outcome was hospital or 30-day mortality. Secondary outcome was acute respiratory distress syndrome (ARDS) or acute lung injury (ALI). Random- or fixed-effect models were taken for quantitative synthesis of the data. RESULTS Antiplatelet therapy was associated with decreased mortality (odds ratio (OR) 0.61; 95% confidence interval (CI), 0.52-0.71; I2 = 0%; P <0. 001) and ARDS/ALI (OR 0.64; 95% CI, 0.50-0.82; I2 = 0%; P <0. 001). In every stratum of subgroups, similar findings on mortality reduction were consistently observed in critically ill patients. CONCLUSIONS Antiplatelet therapy is associated with reduced mortality and lower incidence of ARDS/ALI in critically ill patients, particularly those with predisposing conditions such as high-risk surgery, trauma, pneumonia, and sepsis. However, it remains unclear whether similar findings can be observed in the unselected and broad population with critical illness.
Collapse
Affiliation(s)
- Lijun Wang
- Department of Anesthesiology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Heng Li
- Department of Anesthesiology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Xiaofei Gu
- Department of Anesthesiology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Zhen Wang
- Department of Anesthesiology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Su Liu
- Department of Anesthesiology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Liyong Chen
- Department of Anesthesiology, Daping Hospital, Third Military Medical University, Chongqing, China
- * E-mail:
| |
Collapse
|
41
|
Blockade of Thrombopoietin Reduces Organ Damage in Experimental Endotoxemia and Polymicrobial Sepsis. PLoS One 2016; 11:e0151088. [PMID: 26963510 PMCID: PMC4786277 DOI: 10.1371/journal.pone.0151088] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 02/22/2016] [Indexed: 12/23/2022] Open
Abstract
Background and Purpose Thrombopoietin (TPO), a growth factor primarily involved in thrombopoiesis may also have a role in the pathophysiology of sepsis. In patients with sepsis, indeed, TPO levels are markedly increased, with disease severity being the major independent determinant of TPO concentrations. Moreover, TPO increases and correlates with ex vivo indices of platelet activation in patients with burn injury upon sepsis development, and may contribute to depress cardiac contractility in septic shock. Still, the role of TPO in sepsis pathophysiology remains controversial, given the protective role of TPO in other experimental disease models, for instance in doxorubicin-induced cardiotoxicity and myocardial ischemia/reperfusion injury. The aim of our study was to define the contribution of TPO in the development of organ damage induced by endotoxemia or sepsis, and to investigate the effects of inhibiting TPO in these conditions. Methods We synthesized a chimeric protein able to inhibit TPO, mTPOR-MBP, and studied its effect in two murine experimental models, acute endotoxemia and cecal ligation and puncture (CLP) model. Results In both models, TPO levels markedly increased, from 289.80±27.87 pg/mL to 465.60±45.92 pg/mL at 3 hours in the LPS model (P<0.01), and from 265.00±26.02 pg/mL to 373.70±26.20 pg/mL in the CLP model (P<0.05), respectively. Paralleling TPO levels, also platelet-monocyte aggregates increased, from 32.86±2.48% to 46.13±1.39% at 3 hours in the LPS model (P<0.01), and from 43.68±1.69% to 56.52±4.66% in the CLP model (P<0.05). Blockade of TPO by mTPOR-MBP administration reduced histological damage in target organs, namely lung, liver, and gut. In particular, neutrophil infiltration and lung septal thickening were reduced from a score of 1.86±0.34 to 0.60±0.27 (P<0.01) and from 1.43±0.37 to 0.40±0.16 (P<0.05), respectively, in the LPS model at 3 hours, and from a score of 1.75±0.37 to 0.38±0.18 (P<0.01) and from 1.25±0.31 to 0.13±0.13 (P<0.001), respectively, in the CLP model. Similarly, the number of hepatic microabscesses was decreased from 14.14±1.41 to 3.64±0.56 in the LPS model at 3 hours (P<0.001), and from 1.71±0.29 to 0.13±0.13 in the CLP model (P<0.001). Finally, the diameter of intestinal villi decreased from 90.69±3.95 μm to 70.74±3.60 μm in the LPS model at 3 hours (P<0.01), and from 74.29±4.29 μm to 57.50±1.89 μm in the CLP model (P<0.01). This protective effect was associated with the blunting of the increase in platelet-monocyte adhesion, and, on the contrary, with increased platelet-neutrophil aggregates in the circulation, which may be related to decreased neutrophil sequestration into the inflamed tissues. Conversely, circulating cytokine levels were not significantly changed, in both models, by mTPOR-MBP administration. Conclusion Our results demonstrate that TPO participates in the development of organ damage induced by experimental endotoxemia or polymicrobial sepsis via a mechanism involving increased platelet-leukocyte adhesion, but not cytokine release, and suggest that blocking TPO may be useful in preventing organ damage in patients affected by systemic inflammatory response or sepsis.
Collapse
|
42
|
Al Harbi SA, Tamim HM, Al-Dorzi HM, Sadat M, Arabi YM. Association between aspirin therapy and the outcome in critically ill patients: a nested cohort study. BMC Pharmacol Toxicol 2016; 17:5. [PMID: 26850706 PMCID: PMC4743206 DOI: 10.1186/s40360-016-0047-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Accepted: 01/14/2016] [Indexed: 12/25/2022] Open
Abstract
Background Antiplatelet therapy may attenuate the undesirable effects of platelets on the inflammatory cascades in critical illness. The objective of this study was to evaluate the association between aspirin therapy during intensive care unit (ICU) stay and all-cause mortality. Methods This was a nested cohort study within two randomized controlled trials in which all enrolled patients (N = 763) were grouped according to aspirin intake during ICU stay. The primary endpoints were all-cause ICU mortality and hospital mortality. Secondary endpoints included the development of severe sepsis during the ICU stay, ICU and hospital length of stay and the duration of mechanical ventilation. Propensity score was used to adjust for clinically and statistically relevant variables. Results Of the 763 patients, 154 patients (20 %) received aspirin. Aspirin therapy was not associated with a reduction in ICU mortality (adjusted OR 1.18, 95 % CI 0.69–2.02, P = 0.55) nor with hospital mortality (adjusted OR 0.95, 95 % CI 0.61–1.50, P = 0.82). Aspirin use had no preferential association with mortality among any of the study subgroups. Additionally, aspirin therapy was associated with higher risk of ICU-acquired severe sepsis, and increased mechanical ventilation duration and ICU length of stay. Conclusion Our study showed that the use of aspirin in critically ill patients was not associated with lower mortality, but rather with an increased morbidity. Trial Registration Number ISRCTN07413772 and ISRCTN96294863.
Collapse
Affiliation(s)
- Shmeylan A Al Harbi
- Pharmaceutical Care Department, College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Hani M Tamim
- Department of Internal Medicine, American University of Beirut-Medical Center, Beirut, Lebanon
| | - Hasan M Al-Dorzi
- Intensive Care Department, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, PO Box 22490, MC 1425, Riyadh, 1426 K S A
| | | | - Yaseen M Arabi
- Intensive Care Department, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, PO Box 22490, MC 1425, Riyadh, 1426 K S A.
| |
Collapse
|
43
|
Svensson L, Frick IM, Shannon O. Group G streptococci mediate fibrinogen-dependent platelet aggregation leading to transient entrapment in platelet aggregates. MICROBIOLOGY-SGM 2015; 162:117-126. [PMID: 26511072 DOI: 10.1099/mic.0.000203] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Platelets have been reported to become activated in response to bacteria and this is proposed to contribute to the acute response to bacterial infection. In the present study, we investigated platelet aggregation in response to group G streptococci (GGS) in vitro in healthy human donors and in vivo in a mouse model of streptococcal sepsis. Platelet aggregation by GGS was dependent on the bacterial surface protein FOG and engagement of the platelet fibrinogen receptor; however, it was independent of IgG and the platelet Fc receptor. Platelets exerted no antibacterial effects on the bacteria, and aggregates formed were markedly unstable, allowing bacteria to rapidly return to the plasma and grow post-aggregation. Thrombocytopenia and platelet activation occurred during invasive infection with GGS, and platelets were demonstrated to contribute to bacterial dissemination during infection. These findings reveal an important role for bacteria-platelet interactions during the pathogenesis of streptococcal infection.
Collapse
Affiliation(s)
- Lisbeth Svensson
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, SE-22184 Lund, Sweden
| | - Inga-Maria Frick
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, SE-22184 Lund, Sweden
| | - Oonagh Shannon
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, SE-22184 Lund, Sweden
| |
Collapse
|
44
|
Mederle K, Meurer M, Castrop H, Höcherl K. Inhibition of COX-1 attenuates the formation of thromboxane A2 and ameliorates the acute decrease in glomerular filtration rate in endotoxemic mice. Am J Physiol Renal Physiol 2015; 309:F332-40. [DOI: 10.1152/ajprenal.00567.2014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 05/11/2015] [Indexed: 11/22/2022] Open
Abstract
Thromboxane (Tx) A2 has been suggested to be involved in the development of sepsis-induced acute kidney injury (AKI). Therefore, we investigated the impact of cyclooxygenase (COX)-1 and COX-2 activity on lipopolysaccharide (LPS)-induced renal TxA2 formation, and on endotoxemia-induced AKI in mice. Injection of LPS (3 mg/kg ip) decreased glomerular filtration rate (GFR) and the amount of thrombocytes to ∼50% of basal values after 4 h. Plasma and renocortical tissue levels of TxB2 were increased ∼10- and 1.7-fold in response to LPS, respectively. The COX-1 inhibitor SC-560 attenuated the LPS-induced fall in GFR and in platelet count to ∼75% of basal levels. Furthermore, SC-560 abolished the increase in plasma and renocortical tissue levels of TxB2 in response to LPS. The COX-2 inhibitor SC-236 further enhanced the LPS-induced decrease in GFR to ∼40% of basal values. SC-236 did not alter thrombocyte levels nor the LPS-induced increase in plasma and renocortical tissue levels of TxB2. Pretreatment with clopidogrel inhibited the LPS-induced drop in thrombocyte count, but did not attenuate the LPS-induced decrease in GFR and the increase in plasma TxB2 levels. This study demonstrates that endotoxemia-induced TxA2 formation depends on the activity of COX-1. Our study further indicates that the COX-1 inhibitor SC-560 has a protective effect on the decrease in renal function in response to endotoxin. Therefore, our data support a role for TxA2 in the development of AKI in response to LPS.
Collapse
Affiliation(s)
- Katharina Mederle
- Institute of Physiology, University of Regensburg, Regensburg, Germany; and
| | - Manuel Meurer
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Hayo Castrop
- Institute of Physiology, University of Regensburg, Regensburg, Germany; and
| | - Klaus Höcherl
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
45
|
Thomas MR, Storey RF. Effect of P2Y12 inhibitors on inflammation and immunity. Thromb Haemost 2015; 114:490-7. [PMID: 26156883 DOI: 10.1160/th14-12-1068] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 05/04/2015] [Indexed: 02/04/2023]
Abstract
Platelet P2Y12 inhibitors form a major part of the treatment strategy for patients with acute coronary syndromes (ACS) due to the importance of the platelet P2Y12 receptor in mediating the pathophysiology of arterial thrombosis. It has been increasingly recognised that platelets also have a critical role in inflammation and immune responses. P2Y12 inhibitors reduce platelet release of pro-inflammatory α-granule contents and the formation of pro-inflammatory platelet-leukocyte aggregates. These are important mediators of inflammation in a variety of different contexts. Clinical evidence shows that P2Y12 inhibition by clopidogrel is associated with a reduction in platelet-related mediators of inflammation, such as soluble P-selectin and CD40L, following atherothrombosis. Clopidogrel in addition to aspirin, compared to aspirin alone, also reduces markers of systemic inflammation such as tumour necrosis factor (TNF) α and C-reactive protein (CRP) following ACS. The more potent thienopyridine P2Y12 inhibitor, prasugrel, has been shown to decrease platelet P-selectin expression and platelet-leukocyte aggregate formation compared to clopidogrel. The PLATO study suggested that the novel P2Y12 inhibitor ticagrelor might improve clinical outcomes from pulmonary infections and sepsis compared to clopidogrel in patients with ACS. Ticagrelor is a more potent P2Y12 inhibitor than clopidogrel and also inhibits cellular adenosine uptake via equilibrative nucleoside transporter (ENT) 1, whereas clopidogrel does not. Further examination of the involvement of these mechanisms in inflammation and immunity is therefore warranted.
Collapse
Affiliation(s)
- Mark R Thomas
- Dr. Mark R. Thomas, BMedSci BMBS MRCP, Department of Cardiovascular Science, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK, Tel.: +44 114 3052019, Fax: +44 114 2266159, E-mail
| | | |
Collapse
|
46
|
Abstract
OBJECTIVES Streptococcus pneumoniae is the most common causative pathogen in community-acquired pneumonia. In patients, thrombocytopenia is correlated with an adverse outcome of pneumonia. Platelets can modulate the host response to infection in several ways, that is, by facilitating clot formation, production of antimicrobial proteins, and interaction with neutrophils. We studied the effect of thrombocytopenia during murine pneumococcal pneumonia. DESIGN Animal study. SETTING University research laboratory. SUBJECTS Mice. INTERVENTIONS Pneumonia was induced by intranasal inoculation of S. pneumoniae. Platelets were depleted by anti-mouse thrombocyte serum; controls received nonimmunogenic serum. In separate studies, mice were treated with the platelet P2Y12 receptor inhibitor clopidogrel or placebo. MEASUREMENTS AND MAIN RESULTS Thrombocytopenic mice (platelet counts < 1% of uninfected controls) showed a reduced survival during pneumococcal pneumonia (27% vs 75% among controls; p = 0.003), which was associated with higher bacterial loads in lungs, spleen, and blood. Thrombocytopenic mice showed enhanced coagulation activation (thrombin-antithrombin complexes) in plasma. Proinflammatory cytokine levels were higher in plasma but not in lungs of thrombocytopenic mice. Although clopidogrel treatment strongly prolonged the bleeding time, it did not impact on bacterial loads during pneumococcal pneumonia. CONCLUSIONS Platelets play a protective role during pneumococcal pneumonia independent of their aggregation.
Collapse
|
47
|
Tsai MJ, Ou SM, Shih CJ, Chao PW, Wang LF, Shih YN, Li SY, Kuo SC, Hsu YT, Chen YT. Association of prior antiplatelet agents with mortality in sepsis patients: a nationwide population-based cohort study. Intensive Care Med 2015; 41:806-13. [PMID: 25829229 DOI: 10.1007/s00134-015-3760-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 03/16/2015] [Indexed: 01/08/2023]
Abstract
BACKGROUND Antiplatelet agents are widely used for cardiovascular disea ses, but their pleiotropic effects in sepsis are controversial. OBJECTIVE To investigate the association between antiplatelet agents and the survival benefit for sepsis patients. DESIGN A nationwide population-based cohort and nested case-control study. SETTING Taiwan National Health Insurance database. PARTICIPANTS All patients (age ≥18 years) who were hospitalized for sepsis between January 2000 and December 2010. MEASUREMENTS Conditional logistic regression was used to adjust for confounding. Adjusted odd ratios (ORs) were used to compare the mortality rate due to sepsis in antiplatelet drug users and nonusers. RESULTS Of 683,421 included patients, 229,792 (33.6 %) patients died during hospitalization for sepsis, and the rest (64.4 %) survived to discharge. Use of antiplatelet agents before admission was associated with a lower risk of mortality in sepsis patients (aOR 0.82, 95 % confidence interval [CI] 0.81-0.83, P < 0.001). By using another case-control study design, the beneficial effect was more significant in current users (aOR 0.78, 95 % CI 0.76-0.79) than in recent users (aOR 0.88, 95 % CI 0.85-0.91), but was not significant in past users (aOR 1.00, 95 % CI 0.98-1.02). LIMITATIONS Observational study. CONCLUSIONS Prior use of antiplatelet agents was associated with a survival benefit in sepsis patients.
Collapse
Affiliation(s)
- Min-Juei Tsai
- Department of Internal Medicine, Taipei Veterans General Hospital, Suao Branch, Yilan, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Boyle AJ, Di Gangi S, Hamid UI, Mottram LJ, McNamee L, White G, Cross LJM, McNamee JJ, O'Kane CM, McAuley DF. Aspirin therapy in patients with acute respiratory distress syndrome (ARDS) is associated with reduced intensive care unit mortality: a prospective analysis. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2015; 19:109. [PMID: 25887566 PMCID: PMC4371625 DOI: 10.1186/s13054-015-0846-4] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 03/02/2015] [Indexed: 02/07/2023]
Abstract
Introduction Acute respiratory distress syndrome (ARDS) is a common clinical syndrome with high mortality and long-term morbidity. To date there is no effective pharmacological therapy. Aspirin therapy has recently been shown to reduce the risk of developing ARDS, but the effect of aspirin on established ARDS is unknown. Methods In a single large regional medical and surgical ICU between December 2010 and July 2012, all patients with ARDS were prospectively identified and demographic, clinical, and laboratory variables were recorded retrospectively. Aspirin usage, both pre-hospital and during intensive care unit (ICU) stay, was included. The primary outcome was ICU mortality. We used univariate and multivariate logistic regression analyses to assess the impact of these variables on ICU mortality. Results In total, 202 patients with ARDS were included; 56 (28%) of these received aspirin either pre-hospital, in the ICU, or both. Using multivariate logistic regression analysis, aspirin therapy, given either before or during hospital stay, was associated with a reduction in ICU mortality (odds ratio (OR) 0.38 (0.15 to 0.96) P = 0.04). Additional factors that predicted ICU mortality for patients with ARDS were vasopressor use (OR 2.09 (1.05 to 4.18) P = 0.04) and APACHE II score (OR 1.07 (1.02 to 1.13) P = 0.01). There was no effect upon ICU length of stay or hospital mortality. Conclusion Aspirin therapy was associated with a reduced risk of ICU mortality. These data are the first to demonstrate a potential protective role for aspirin in patients with ARDS. Clinical trials to evaluate the role of aspirin as a pharmacological intervention for ARDS are needed.
Collapse
Affiliation(s)
- Andrew J Boyle
- Centre for Infection and Immunity, Health Sciences Building, Queen's University Belfast, 97 Lisburn Road, Belfast, UK. .,Regional Intensive Care Unit, Royal Victoria Hospital, Grosvenor Road, Belfast, UK.
| | - Stefania Di Gangi
- Epidemiology ASL TO3, Via Sabaudia, 164, Grugliasco, TO, 10095, Italy.
| | - Umar I Hamid
- Centre for Infection and Immunity, Health Sciences Building, Queen's University Belfast, 97 Lisburn Road, Belfast, UK.
| | - Linda-Jayne Mottram
- Regional Intensive Care Unit, Royal Victoria Hospital, Grosvenor Road, Belfast, UK.
| | - Lia McNamee
- Regional Intensive Care Unit, Royal Victoria Hospital, Grosvenor Road, Belfast, UK.
| | - Griania White
- Regional Intensive Care Unit, Royal Victoria Hospital, Grosvenor Road, Belfast, UK.
| | - L J Mark Cross
- Centre for Infection and Immunity, Health Sciences Building, Queen's University Belfast, 97 Lisburn Road, Belfast, UK. .,Regional Intensive Care Unit, Royal Victoria Hospital, Grosvenor Road, Belfast, UK.
| | - James J McNamee
- Regional Intensive Care Unit, Royal Victoria Hospital, Grosvenor Road, Belfast, UK.
| | - Cecilia M O'Kane
- Centre for Infection and Immunity, Health Sciences Building, Queen's University Belfast, 97 Lisburn Road, Belfast, UK.
| | - Daniel F McAuley
- Centre for Infection and Immunity, Health Sciences Building, Queen's University Belfast, 97 Lisburn Road, Belfast, UK. .,Regional Intensive Care Unit, Royal Victoria Hospital, Grosvenor Road, Belfast, UK.
| |
Collapse
|
49
|
Coagulation abnormalities in sepsis. ACTA ACUST UNITED AC 2015; 53:16-22. [DOI: 10.1016/j.aat.2014.11.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 11/16/2014] [Accepted: 11/24/2014] [Indexed: 11/30/2022]
|
50
|
|