Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 237
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
PLoS Biol ; 19(2): e3001109, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33596198

RESUMEN

Coronavirus Disease 2019 (COVID-19), caused by the novel Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), has affected over 30 million globally to date. Although high rates of venous thromboembolism and evidence of COVID-19-induced endothelial dysfunction have been reported, the precise aetiology of the increased thrombotic risk associated with COVID-19 infection remains to be fully elucidated. Therefore, we assessed clinical platelet parameters and circulating platelet activity in patients with severe and nonsevere COVID-19. An assessment of clinical blood parameters in patients with severe COVID-19 disease (requiring intensive care), patients with nonsevere disease (not requiring intensive care), general medical in-patients without COVID-19, and healthy donors was undertaken. Platelet function and activity were also assessed by secretion and specific marker analysis. We demonstrated that routine clinical blood parameters including increased mean platelet volume (MPV) and decreased platelet:neutrophil ratio are associated with disease severity in COVID-19 upon hospitalisation and intensive care unit (ICU) admission. Strikingly, agonist-induced ADP release was 30- to 90-fold higher in COVID-19 patients compared with hospitalised controls and circulating levels of platelet factor 4 (PF4), soluble P-selectin (sP-selectin), and thrombopoietin (TPO) were also significantly elevated in COVID-19. This study shows that distinct differences exist in routine full blood count and other clinical laboratory parameters between patients with severe and nonsevere COVID-19. Moreover, we have determined all COVID-19 patients possess hyperactive circulating platelets. These data suggest abnormal platelet reactivity may contribute to hypercoagulability in COVID-19 and confirms the role that platelets/clotting has in determining the severity of the disease and the complexity of the recovery path.


Asunto(s)
Plaquetas/virología , COVID-19/sangre , Adenosina Trifosfato/metabolismo , Anciano , Coagulación Sanguínea , Plaquetas/citología , Ensayo de Inmunoadsorción Enzimática , Femenino , Hemostasis , Humanos , Inflamación , Unidades de Cuidados Intensivos , Masculino , Volúmen Plaquetario Medio , Persona de Mediana Edad , Selectina-P/sangre , Fenotipo , Factor Plaquetario 4/sangre , Pruebas de Función Plaquetaria , Trombopoyetina/sangre
2.
PLoS Pathog ; 17(5): e1009587, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33974679

RESUMEN

Severe fever with thrombocytopenia syndrome virus (SFTSV) is a tick-borne emerging phlebovirus with high mortality rates of 6.0 to 30%. SFTSV infection is characterized by high fever, thrombocytopenia, leukopenia, hemorrhage and multiple organ failures. Currently, specific therapies and vaccines remain elusive. Suitable small animal models are urgently needed to elucidate the pathogenesis and evaluate the potential drug and vaccine for SFTSV infection. Previous models presented only mild or no pathogenesis of SFTS, limiting their applications in SFTSV infection. Therefore, it is an urgent need to develop a small animal model for the investigation of SFTSV pathogenesis and evaluation of therapeutics. In the current report, we developed a SFTSV infection model based on the HuPBL-NCG mice that recapitulates many pathological characteristics of SFTSV infection in humans. Virus-induced histopathological changes were identified in spleen, lung, kidney, and liver. SFTSV was colocalized with macrophages in the spleen and liver, suggesting that the macrophages in the spleen and liver could be the principle target cells of SFTSV. In addition, histological analysis showed that the vascular endothelium integrity was severely disrupted upon viral infection along with depletion of platelets. In vitro cellular assays further revealed that SFTSV infection increased the vascular permeability of endothelial cells by promoting tyrosine phosphorylation and internalization of the adhesion molecule vascular endothelial (VE)-cadherin, a critical component of endothelial integrity. In addition, we found that both virus infection and pathogen-induced exuberant cytokine release dramatically contributed to the vascular endothelial injury. We elucidated the pathogenic mechanisms of hemorrhage syndrome and developed a humanized mouse model for SFTSV infection, which should be helpful for anti-SFTSV therapy and pathogenesis study.


Asunto(s)
Modelos Animales de Enfermedad , Phlebovirus/patogenicidad , Síndrome de Trombocitopenia Febril Grave/patología , Enfermedades por Picaduras de Garrapatas/patología , Animales , Plaquetas/patología , Plaquetas/virología , Moléculas de Adhesión Celular/metabolismo , Células Endoteliales/patología , Células Endoteliales/virología , Femenino , Humanos , Leucocitos Mononucleares/patología , Leucocitos Mononucleares/virología , Macrófagos/patología , Macrófagos/virología , Ratones , Fosforilación , Síndrome de Trombocitopenia Febril Grave/virología , Enfermedades por Picaduras de Garrapatas/virología
4.
Blood ; 136(11): 1317-1329, 2020 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-32573711

RESUMEN

There is an urgent need to understand the pathogenesis of coronavirus disease 2019 (COVID-19). In particular, thrombotic complications in patients with COVID-19 are common and contribute to organ failure and mortality. Patients with severe COVID-19 present with hemostatic abnormalities that mimic disseminated intravascular coagulopathy associated with sepsis, with the major difference being increased risk of thrombosis rather than bleeding. However, whether severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection alters platelet function to contribute to the pathophysiology of COVID-19 remains unknown. In this study, we report altered platelet gene expression and functional responses in patients infected with SARS-CoV-2. RNA sequencing demonstrated distinct changes in the gene-expression profile of circulating platelets of COVID-19 patients. Pathway analysis revealed differential gene-expression changes in pathways associated with protein ubiquitination, antigen presentation, and mitochondrial dysfunction. The receptor for SARS-CoV-2 binding, angiotensin-converting enzyme 2 (ACE2), was not detected by messenger RNA (mRNA) or protein in platelets. Surprisingly, mRNA from the SARS-CoV-2 N1 gene was detected in platelets from 2 of 25 COVID-19 patients, suggesting that platelets may take-up SARS-COV-2 mRNA independent of ACE2. Resting platelets from COVID-19 patients had increased P-selectin expression basally and upon activation. Circulating platelet-neutrophil, -monocyte, and -T-cell aggregates were all significantly elevated in COVID-19 patients compared with healthy donors. Furthermore, platelets from COVID-19 patients aggregated faster and showed increased spreading on both fibrinogen and collagen. The increase in platelet activation and aggregation could partially be attributed to increased MAPK pathway activation and thromboxane generation. These findings demonstrate that SARS-CoV-2 infection is associated with platelet hyperreactivity, which may contribute to COVID-19 pathophysiology.


Asunto(s)
Betacoronavirus/aislamiento & purificación , Trastornos de la Coagulación Sanguínea/patología , Plaquetas/patología , Infecciones por Coronavirus/complicaciones , Neumonía Viral/complicaciones , Transcriptoma , Biomarcadores , Trastornos de la Coagulación Sanguínea/genética , Trastornos de la Coagulación Sanguínea/metabolismo , Trastornos de la Coagulación Sanguínea/virología , Plaquetas/metabolismo , Plaquetas/virología , COVID-19 , Estudios de Casos y Controles , Infecciones por Coronavirus/genética , Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/virología , Femenino , Estudios de Seguimiento , Perfilación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Pandemias , Neumonía Viral/genética , Neumonía Viral/metabolismo , Neumonía Viral/virología , Pronóstico , Estudios Prospectivos , SARS-CoV-2
5.
Blood ; 136(11): 1330-1341, 2020 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-32678428

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an emergent pathogen responsible for the coronavirus disease 2019 (COVID-19). Since its emergence, the novel coronavirus has rapidly achieved pandemic proportions causing remarkably increased morbidity and mortality around the world. A hypercoagulability state has been reported as a major pathologic event in COVID-19, and thromboembolic complications listed among life-threatening complications of the disease. Platelets are chief effector cells of hemostasis and pathological thrombosis. However, the participation of platelets in the pathogenesis of COVID-19 remains elusive. This report demonstrates that increased platelet activation and platelet-monocyte aggregate formation are observed in severe COVID-19 patients, but not in patients presenting mild COVID-19 syndrome. In addition, exposure to plasma from severe COVID-19 patients increased the activation of control platelets ex vivo. In our cohort of COVID-19 patients admitted to the intensive care unit, platelet-monocyte interaction was strongly associated with tissue factor (TF) expression by the monocytes. Platelet activation and monocyte TF expression were associated with markers of coagulation exacerbation as fibrinogen and D-dimers, and were increased in patients requiring invasive mechanical ventilation or patients who evolved with in-hospital mortality. Finally, platelets from severe COVID-19 patients were able to induce TF expression ex vivo in monocytes from healthy volunteers, a phenomenon that was inhibited by platelet P-selectin neutralization or integrin αIIb/ß3 blocking with the aggregation inhibitor abciximab. Altogether, these data shed light on new pathological mechanisms involving platelet activation and platelet-dependent monocyte TF expression, which were associated with COVID-19 severity and mortality.


Asunto(s)
Betacoronavirus/inmunología , Trastornos de la Coagulación Sanguínea/patología , Plaquetas/patología , Infecciones por Coronavirus/complicaciones , Monocitos/patología , Neumonía Viral/complicaciones , Tromboplastina/metabolismo , Adulto , Biomarcadores/metabolismo , Trastornos de la Coagulación Sanguínea/inmunología , Trastornos de la Coagulación Sanguínea/metabolismo , Trastornos de la Coagulación Sanguínea/virología , Plaquetas/metabolismo , Plaquetas/virología , COVID-19 , Estudios de Casos y Controles , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/virología , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Monocitos/metabolismo , Monocitos/virología , Selectina-P/metabolismo , Pandemias , Activación Plaquetaria , Neumonía Viral/inmunología , Neumonía Viral/metabolismo , Neumonía Viral/virología , Pronóstico , Estudios Prospectivos , SARS-CoV-2 , Tasa de Supervivencia
6.
J Biomed Sci ; 29(1): 52, 2022 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-35820906

RESUMEN

BACKGROUND: Coronavirus-induced disease 19 (COVID-19) infects more than three hundred and sixty million patients worldwide, and people with severe symptoms frequently die of acute respiratory distress syndrome (ARDS). Recent studies indicated that excessive neutrophil extracellular traps (NETs) contributed to immunothrombosis, thereby leading to extensive intravascular coagulopathy and multiple organ dysfunction. Thus, understanding the mechanism of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced NET formation would be helpful to reduce thrombosis and prevent ARDS in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. METHODS: We incubated SARS-CoV-2 with neutrophils in the presence or absence of platelets to observe NET formation. We further isolated extracellular vesicles from COVID-19 patients' sera (COVID-19-EVs) to examine their ability to induce NET formation. RESULTS: We demonstrated that antagonistic mAbs against anti-CLEC5A mAb and anti-TLR2 mAb can inhibit COVID-19-EVs-induced NET formation, and generated clec5a-/-/tlr2-/- mice to confirm the critical roles of CLEC5A and TLR2 in SARS-CoV-2-induced lung inflammation in vivo. We found that virus-free extracellular COVID-19 EVs induced robust NET formation via Syk-coupled C-type lectin member 5A (CLEC5A) and TLR2. Blockade of CLEC5A inhibited COVID-19 EVs-induced NETosis, and simultaneous blockade of CLEC5A and TLR2 further suppressed SARS-CoV-2-induced NETosis in vitro. Moreover, thromboinflammation was attenuated dramatically in clec5a-/-/tlr2-/- mice. CONCLUSIONS: This study demonstrates that SARS-CoV-2-activated platelets produce EVs to enhance thromboinflammation via CLEC5A and TLR2, and highlight the importance of CLEC5A and TLR2 as therapeutic targets to reduce the risk of ARDS in COVID-19 patients.


Asunto(s)
COVID-19 , Lectinas Tipo C , Neutrófilos , Neumonía , Síndrome de Dificultad Respiratoria , SARS-CoV-2 , Trombosis , Animales , Plaquetas/inmunología , Plaquetas/patología , Plaquetas/virología , COVID-19/sangre , COVID-19/inmunología , Humanos , Lectinas Tipo C/inmunología , Ratones , Neutrófilos/inmunología , Neutrófilos/patología , Neutrófilos/virología , Neumonía/inmunología , Neumonía/patología , Neumonía/virología , Receptores de Superficie Celular , Síndrome de Dificultad Respiratoria/inmunología , Síndrome de Dificultad Respiratoria/virología , SARS-CoV-2/inmunología , Trombosis/sangre , Trombosis/inmunología , Trombosis/virología , Receptor Toll-Like 2/inmunología
7.
Platelets ; 33(1): 48-53, 2022 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-34847829

RESUMEN

Coagulopathy is an evident complication of COVID-19 with predominance of a prothrombotic state. Platelet activation plays a key role. The terms "hyper-reactivity" and "hyperactivity" used in recent literature may not be clear or sufficient to explain the pathological events involved in COVID-related thrombosis (CRT). Inflammation may play a bigger role compared to thrombosis in COVID-related mortality because a smaller percentage of patients with COVID-19 die due to direct effects of thrombosis. Not all COVID-19 patients have thrombocytopenia and a few show thrombocytosis. We believe the platelet pathology is more complex than just activation or hyper-activation, particularly due to the platelets' role in inflammation. Understanding the pathology and consequences of platelets' role may help optimize management strategies and diminish CRT-associated morbidity and mortality. In this viewpoint report, we examine the published evidence of platelet hyper-reactivity in COVID-19 with a focused analysis of the key pathologies, diverse alterations, disease outcomes, and therapeutic targets. We believe that COVID-19 is a disease of inflammation and pathologic platelets, and based on the complexity and diverse pathologies, we propose the term "thrombocytopathy" as a more reflective term of the platelets' involvement in COVID-19. In our opinion, thrombocytopathy is the unpredictable pathologic alterations of platelets in function, morphology and number, caused by different factors with a variety of presentations.


Asunto(s)
Plaquetas/patología , COVID-19/complicaciones , Síndrome de Liberación de Citoquinas/complicaciones , Coagulación Intravascular Diseminada/complicaciones , Embolia Pulmonar/complicaciones , SARS-CoV-2/patogenicidad , Abciximab/uso terapéutico , Enfermedad Aguda , Anticoagulantes/uso terapéutico , Aspirina/uso terapéutico , Plaquetas/efectos de los fármacos , Plaquetas/virología , COVID-19/diagnóstico , COVID-19/virología , Clopidogrel/uso terapéutico , Síndrome de Liberación de Citoquinas/diagnóstico , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Síndrome de Liberación de Citoquinas/virología , Coagulación Intravascular Diseminada/diagnóstico , Coagulación Intravascular Diseminada/tratamiento farmacológico , Coagulación Intravascular Diseminada/virología , Fibrinolíticos/uso terapéutico , Humanos , Pulmón/irrigación sanguínea , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/virología , Activación Plaquetaria/efectos de los fármacos , Embolia Pulmonar/diagnóstico , Embolia Pulmonar/tratamiento farmacológico , Embolia Pulmonar/virología , Resultado del Tratamiento , Tratamiento Farmacológico de COVID-19
8.
J Med Virol ; 93(9): 5425-5431, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33945642

RESUMEN

A rapid outbreak of novel coronavirus, coronavirus disease-2019 (COVID-19), has made it a global pandemic. This study focused on the possible association between lymphopenia and computed tomography (CT) scan features and COVID-19 patient mortality. The clinical data of 596 COVID-19 patients were collected from February 2020 to September 2020. The patients' serological survey and CT scan features were retrospectively explored. The median age of the patients was 56.7 ± 16.4 years old. Lung involvement was more than 50% in 214 COVID-19 patients (35.9%). The average blood lymphocyte percentage was 20.35 ± 10.16 (normal range, 20%-50%). Although the levels of C-reactive protein (CRP) and erythrocyte sedimentation rate (ESR) were high in more than 80% of COVID-19 patients; CRP, ESR, and platelet-to-lymphocyte ratio (PLR) may not indicate the in-hospital mortality of COVID-19. Patients with severe lung involvement and lymphopenia were found to be significantly associated with increased odds of death (odds ratio, 9.24; 95% confidence interval, 4.32-19.78). These results indicated that lymphopenia < 20% along with pulmonary involvement >50% impose a multiplicative effect on the risk of mortality. The in-hospital mortality rate of this group was significantly higher than other COVID-19 hospitalized cases. Furthermore, they meaningfully experienced a prolonged stay in the hospital (p = .00). Lymphocyte count less than 20% and chest CT scan findings with more than 50% involvement might be related to the patient's mortality. These could act as laboratory and clinical indicators of disease severity, mortality, and outcome.


Asunto(s)
COVID-19/complicaciones , Pulmón/patología , Linfopenia/complicaciones , Neumonía/complicaciones , SARS-CoV-2/patogenicidad , Adulto , Anciano , Biomarcadores/sangre , Plaquetas/patología , Plaquetas/virología , Sedimentación Sanguínea , Proteína C-Reactiva , COVID-19/diagnóstico por imagen , COVID-19/mortalidad , COVID-19/virología , Femenino , Mortalidad Hospitalaria , Humanos , Irán , Pulmón/virología , Linfocitos/patología , Linfocitos/virología , Linfopenia/diagnóstico por imagen , Linfopenia/mortalidad , Linfopenia/virología , Masculino , Persona de Mediana Edad , Neumonía/diagnóstico por imagen , Neumonía/mortalidad , Neumonía/virología , Estudios Retrospectivos , Índice de Severidad de la Enfermedad , Análisis de Supervivencia , Tomografía Computarizada por Rayos X
9.
J Med Virol ; 93(9): 5390-5395, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33913549

RESUMEN

Hypercoagulability and thrombosis caused by coronavirus disease 2019 (COVID-19) are related to the higher mortality rate. Because of limited data on the antiplatelet effect, we aimed to evaluate the impact of aspirin add-on therapy on the outcome of the patients hospitalized due to severe COVID-19. In this cohort study, patients with a confirmed diagnosis of severe COVID-19 admitted to Imam Hossein Medical Center, Tehran, Iran from March 2019 to July 2020 were included. Demographics and related clinical data during their hospitalization were recorded. The mortality rate of the patients was considered as the primary outcome and its association with aspirin use was assessed. Nine hundred and ninety-one patients were included, of that 336 patients (34%) received aspirin during their hospitalization and 655 ones (66%) did not. Comorbidities were more prevalent in the patients who were receiving aspirin. Results from the multivariate COX proportional model demonstrated a significant independent association between aspirin use and reduction in the risk of in-hospital mortality (0.746 [0.560-0.994], p = 0.046). Aspirin use in hospitalized patients with COVID-19 is associated with a significant decrease in mortality rate. Further prospective randomized controlled trials are needed to assess the efficacy and adverse effects of aspirin administration in this population.


Asunto(s)
Aspirina/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Coagulación Intravascular Diseminada/tratamiento farmacológico , Inhibidores de Agregación Plaquetaria/uso terapéutico , Embolia Pulmonar/tratamiento farmacológico , SARS-CoV-2/patogenicidad , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/uso terapéutico , Adulto , Anciano , Alanina/análogos & derivados , Alanina/uso terapéutico , Antivirales/uso terapéutico , Plaquetas/efectos de los fármacos , Plaquetas/patología , Plaquetas/virología , COVID-19/complicaciones , COVID-19/mortalidad , COVID-19/virología , Enfermedad de la Arteria Coronaria/complicaciones , Enfermedad de la Arteria Coronaria/tratamiento farmacológico , Enfermedad de la Arteria Coronaria/mortalidad , Enfermedad de la Arteria Coronaria/virología , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/mortalidad , Diabetes Mellitus/virología , Coagulación Intravascular Diseminada/complicaciones , Coagulación Intravascular Diseminada/mortalidad , Coagulación Intravascular Diseminada/virología , Combinación de Medicamentos , Femenino , Mortalidad Hospitalaria , Humanos , Hipertensión/complicaciones , Hipertensión/tratamiento farmacológico , Hipertensión/mortalidad , Hipertensión/virología , Irán , Lopinavir/uso terapéutico , Pulmón/irrigación sanguínea , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/virología , Masculino , Persona de Mediana Edad , Embolia Pulmonar/complicaciones , Embolia Pulmonar/mortalidad , Embolia Pulmonar/virología , Respiración Artificial/mortalidad , Respiración Artificial/estadística & datos numéricos , Estudios Retrospectivos , Ritonavir/uso terapéutico , SARS-CoV-2/efectos de los fármacos , Índice de Severidad de la Enfermedad , Análisis de Supervivencia , Resultado del Tratamiento
10.
Scand J Immunol ; 94(5): e13097, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34940978

RESUMEN

COVID-19 is a global pandemic with a daily increasing number of affected individuals. Thrombosis is a severe complication of COVID-19 that leads to a worse clinical course with higher rates of mortality. Multiple lines of evidence suggest that hyperinflammation plays a crucial role in disease progression. This review compiles clinical data of COVID-19 patients who developed thrombotic complications to investigate the possible role of hyperinflammation in inducing hypercoagulation. A systematic literature search was performed using PubMed, Embase, Medline and Scopus to identify relevant clinical studies that investigated thrombotic manifestations and reported inflammatory and coagulation biomarkers in COVID-19 patients. Only 54 studies met our inclusion criteria, the majority of which demonstrated significantly elevated inflammatory markers. In the cohort studies with control, D-dimer was significantly higher in COVID-19 patients with thrombosis as compared to the control. Pulmonary embolism, deep vein thrombosis and strokes were frequently reported which could be attributed to the hyperinflammatory response associated with COVID-19 and/or to the direct viral activation of platelets and endothelial cells, two mechanisms that are discussed in this review. It is recommended that all admitted COVID-19 patients should be assessed for hypercoagulation. Furthermore, several studies have suggested that anticoagulation may be beneficial, especially in hospitalized non-ICU patients. Although vaccines against SARS-CoV-2 have been approved and distributed in several countries, research should continue in the field of prevention and treatment of COVID-19 and its severe complications including thrombosis due to the emergence of new variants against which the efficacy of the vaccines is not yet clear.


Asunto(s)
Arterias/patología , Plaquetas/inmunología , COVID-19/inmunología , Endotelio Vascular/inmunología , Inflamación/inmunología , SARS-CoV-2/fisiología , Trombosis de la Vena/inmunología , Animales , Anticoagulantes/uso terapéutico , Plaquetas/virología , COVID-19/complicaciones , Endotelio Vascular/virología , Humanos , Inflamación/complicaciones , Fenotipo , Trombosis , Trombosis de la Vena/etiología , Trombosis de la Vena/prevención & control
11.
Transfusion ; 61(10): 2885-2897, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34289101

RESUMEN

BACKGROUND: Large volume delayed sampling (LVDS) and pathogen reduction technology (PRT) are strategies for platelet processing to minimize transfusion of contaminated platelet components (PCs). This study holistically compares the economic and clinical impact of LVDS and PRT in the United States. STUDY DESIGN AND METHODS: A decision model was constructed to simulate collection, processing, and use of PCs and to compare processing strategies: PRT with 5-day shelf life, LVDS with 7-day shelf life (LVDS7), and LVDS with 5-day shelf life extended to 7 days with secondary testing (LVDS5/2). Target population was adults requiring two or more transfusions. Collection, processing, storage, and distribution data were obtained from the National Blood Collection and Utilization Survey and published literature. Patient outcomes associated with transfusions were obtained from AABB guidelines, meta-analyses, and other published clinical studies. Costs were obtained from reimbursement schedules and other published sources. RESULTS: Given 10,000 donated units, 9512, 9511, and 9651 units of PRT, LVDS5/2, and LVDS7 PCs were available for transfusion, respectively. With these units, 1502, 2172, and 2329 transfusions can be performed with similar levels of adverse events. Assuming 30 transfusions a day, a hospital would require 69,325, 47,940, and 45,383 units of PRT, LVDS5/2, and LVDS7 platelets to perform these transfusions. The mean costs to perform transfusions were significantly higher with PRT units. CONCLUSIONS: Compared with PRT, LVDS strategies were associated with lower costs and higher PC availability while patients experienced similar levels of adverse events. Increased utilization of LVDS has the potential to improve efficiency, expand patient access to platelets, and reduce health care costs.


Asunto(s)
Plaquetas , Seguridad de la Sangre/métodos , Plaquetas/microbiología , Plaquetas/parasitología , Plaquetas/virología , Seguridad de la Sangre/economía , Humanos , Recuento de Plaquetas , Transfusión de Plaquetas/economía , Transfusión de Plaquetas/métodos , Esterilización/economía , Esterilización/métodos , Estados Unidos
12.
Pediatr Res ; 89(4): 814-822, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32516798

RESUMEN

BACKGROUND: Enterovirus 71 (EV71) infection contributes to hand, foot, and mouth disease (HFMD) with severe neurogenic complications, leading to higher morbidity. In addition to their typical roles in coagulation, platelets could serve as essential immune regulatory cells to play a key role in the pathogenesis of this viral infection. METHODS: Platelet parameters were measured using an automatic hematology analyzer. T-helper type 1 (Th1) and Th2 cells were analyzed by flow cytometry. The levels of cytokines and key transcription factors were determined. RESULTS: The levels of platelet count and plateletcrit were positively associated with the severity of HFMD. Th1 and Th2 cells as well as their corresponding cytokines were increased in the severe group compared to the healthy volunteers. Moreover, the levels of platelets were negatively correlated with the level of interferon-γ (IFN-γ), but positively correlated with the frequency of Th1 cells. Coculture of platelets and naive CD4+ T cells showed that platelets from mild patients promote Th1 cell differentiation and IFN-γ secretion. CONCLUSIONS: Our study has shown for the first time that the distinct roles of platelets are responsible for the regulation of pathogenic CD4+ T cell differentiation and function in the pathogenesis of HFMD caused by EV71. IMPACT: Our study has shown for the first time that the distinct roles of platelets are responsible for the regulation of pathogenic CD4+ T cell differentiation and function in the pathogenesis of HFMD caused by EV71. For the first time, we have discovered the role of platelets in children's HFMD caused by EV71 infection, which may provide a better treatment for HFMD in the future. This article describes new discoveries in platelet immunity.


Asunto(s)
Plaquetas/citología , Plaquetas/virología , Enterovirus Humano A/inmunología , Infecciones por Enterovirus/inmunología , Infecciones por Enterovirus/virología , Enfermedad de Boca, Mano y Pie/inmunología , Enfermedad de Boca, Mano y Pie/virología , Linfocitos T CD4-Positivos/citología , Ligando de CD40/metabolismo , Diferenciación Celular , Niño , Citocinas/metabolismo , Femenino , Voluntarios Sanos , Humanos , Masculino , Selectina-P/metabolismo , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Células TH1/citología , Células Th2/citología
13.
Arterioscler Thromb Vasc Biol ; 40(7): 1635-1650, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32434410

RESUMEN

OBJECTIVE: Thrombocytopenia is associated with many viral infections suggesting virions interact with and affect platelets. Consistently, viral particles are seen inside platelets, and platelet activation markers are detected in viremic patients. In this article, we sought mechanistic insights into these virion/platelet interactions by examining how platelets endocytose, traffic, and are activated by a model virion. Approach and Results: Using fluorescently tagged HIV-1 pseudovirions, 3-dimensional structured illumination microscopy, and transgenic mouse models, we probed the interactions between platelets and virions. Mouse platelets used known endocytic machinery, that is, dynamin, VAMP (vesicle-associated membrane protein)-3, and Arf6 (ADP-ribosylation factor 6), to take up and traffic HIV-1 pseudovirions. Endocytosed HIV-1 pseudovirions trafficked through early (Rab4+) and late endosomes (Rab7+), and then to an LC3+ (microtubule-associated protein 1A/1B-light chain 3) compartment. Incubation with virions induced IRAK4 (interleukin 1 receptor-associated kinase 4), Akt (protein kinase B), and IKK (IκB kinase) activation, granule secretion, and platelet-leukocyte aggregate formation. This activation required TLRs (Toll-like receptors) and MyD88 (myeloid differentiation primary response protein 88) but was less extensive and slower than activation with thrombin. In vivo, HIV-1 pseudovirions injection led to virion uptake and platelet activation, as measured by IKK activation, platelet-leukocyte aggregate formation, and mild thrombocytopenia. All were decreased in VAMP-3-/- and, megakaryocyte/platelet-specific, Arf6-/- mice. Similar platelet activation profiles (increased platelet-leukocyte aggregates, plasma platelet factor 4, and phospho-IκBα) were detected in newly diagnosed and antiretroviral therapy-controlled HIV-1+ patients. CONCLUSIONS: Collectively, our data provide mechanistic insights into the cell biology of how platelets endocytose and process virions. We propose a mechanism by which platelets sample the circulation and respond to potential pathogens that they take up.


Asunto(s)
Plaquetas/metabolismo , Endocitosis , Infecciones por VIH/sangre , VIH-1/patogenicidad , Activación Plaquetaria , Trombocitopenia/sangre , Receptores Toll-Like/sangre , Virión , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/sangre , Factores de Ribosilacion-ADP/genética , Animales , Antirretrovirales/uso terapéutico , Plaquetas/virología , Agregación Celular , Células Cultivadas , Infecciones por VIH/diagnóstico , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/virología , Humanos , Quinasa I-kappa B/sangre , Quinasa I-kappa B/genética , Leucocitos/metabolismo , Leucocitos/virología , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/sangre , Factor 88 de Diferenciación Mieloide/genética , Factor Plaquetario 4/sangre , Factor Plaquetario 4/genética , Trombocitopenia/diagnóstico , Trombocitopenia/virología , Receptores Toll-Like/deficiencia , Receptores Toll-Like/genética , Proteína 3 de Membrana Asociada a Vesículas/sangre , Proteína 3 de Membrana Asociada a Vesículas/genética
14.
Pathobiology ; 88(1): 15-27, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33049751

RESUMEN

Coronavirus disease 2019 (COVID-19) causes a spectrum of disease; some patients develop a severe proinflammatory state which can be associated with a unique coagulopathy and procoagulant endothelial phenotype. Initially, COVID-19 infection produces a prominent elevation of fibrinogen and D-dimer/fibrin(ogen) degradation products. This is associated with systemic hypercoagulability and frequent venous thromboembolic events. The degree of D-dimer elevation positively correlates with mortality in COVID-19 patients. COVID-19 also leads to arterial thrombotic events (including strokes and ischemic limbs) as well as microvascular thrombotic disorders (as frequently documented at autopsy in the pulmonary vascular beds). COVID-19 patients often have mild thrombocytopenia and appear to have increased platelet consumption, together with a corresponding increase in platelet production. Disseminated intravascular coagulopathy (DIC) and severe bleeding events are uncommon in COVID-19 patients. Here, we review the current state of knowledge of COVID-19 and hemostasis.


Asunto(s)
Trastornos de la Coagulación Sanguínea/complicaciones , Plaquetas/virología , COVID-19/virología , SARS-CoV-2/patogenicidad , Trastornos de la Coagulación Sanguínea/etiología , Trastornos de la Coagulación Sanguínea/virología , COVID-19/complicaciones , Productos de Degradación de Fibrina-Fibrinógeno/metabolismo , Humanos , Trombosis/complicaciones
15.
J Thromb Thrombolysis ; 52(1): 105-110, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33387210

RESUMEN

Patients with Coronavirus-associated disease-2019 (COVID-19) display alterations of the hemostatic system and the presence of a prothrombotic status frequently leading to vascular complications. However, the impact of COVID-19 on platelet activity, aggregation and agglutination still needs to be clarified. We measured total levels of von Willebrand factor (vWF) and vWF binding to the platelet glycoprotein (Gp) complex (GPIb-IX-V), in a cohort of COVID-19 patients admitted to the intensive care unit of our Institution. Moreover, we evaluated platelet aggregation in response to agonists (ADP, collagen, arachidonic acid) and platelet agglutination in response to ristocetin. We found that levels of vWF antigen and the active form of vWF binding to platelets (vWF:RCo), were markedly increased in these patients. These results were associated with higher agglutination rates induced by ristocetin, thereby indirectly indicating an increased capability of vWF to bind to platelets. Conversely, we found that platelet aggregation in response to both ADP and collagen was lower in COVID-19 patients compared to healthy volunteers. This study shows that COVID-19 is associated with increased vWF-induced platelet agglutination but reduced platelet responsivity to aggregation stimuli. Our findings have translational relevance since platelet adhesion to vWF may represent a marker to predict possible complications and better delineate therapeutic strategies in COVID-19 patients.


Asunto(s)
Plaquetas/metabolismo , COVID-19/sangre , Agregación Plaquetaria , Factor de von Willebrand/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Aglutinación , Plaquetas/virología , COVID-19/diagnóstico , COVID-19/virología , Femenino , Interacciones Huésped-Patógeno , Humanos , Masculino , Persona de Mediana Edad , Pruebas de Función Plaquetaria , Unión Proteica , SARS-CoV-2/patogenicidad , Trombosis/sangre , Trombosis/diagnóstico , Trombosis/virología
16.
J Thromb Thrombolysis ; 52(3): 708-714, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34519015

RESUMEN

Coronavirus disease 2019 (Covid-19) is associated with a high incidence of venous and arterial thromboembolic events. Currently, there are no clinical or laboratory markers that predict thrombotic risk. Circulating immature platelets are hyper-reactive platelets, which are associated with arterial thrombotic events. The aim of this study was to assess whether the proportion of circulating immature platelets is associated with disease severity in Covid-19 patients. Patients admitted with Covid-19 disease were prospectively assessed. Immature platelet count (IPC) and immature platelet fraction (IPF) were measured at admission and at additional time points during the hospital course using the Sysmex XN-3000 auto-analyzer. A total of 136 consecutive patients with Covid-19 were recruited [mean age 60 ± 19 years, 49% woman, 56 (41%) had mild-moderate disease and 80 (59%) had severe disease at presentation]. The median IPF% was higher in patients with severe compared to mild-moderate disease [5.8 (3.9-8.7) vs. 4.2 (2.73-6.45), respectively, p = 0.01]. The maximal IPC value was also higher in patients with severe disease [15 (10.03-21.56), vs 10.9 (IQR 6.79-15.62), respectively, p = 0.001]. Increased IPC was associated with increased length of hospital stay. Patients with severe Covid-19 have higher levels of IPF than patients with mild-moderate disease. IPF may serve as a prognostic marker for disease severity in Covid-19 patients.


Asunto(s)
Plaquetas/virología , COVID-19/virología , SARS-CoV-2/patogenicidad , Trombosis/virología , Adulto , Anciano , COVID-19/sangre , COVID-19/diagnóstico , COVID-19/terapia , Femenino , Mortalidad Hospitalaria , Interacciones Huésped-Patógeno , Humanos , Tiempo de Internación , Masculino , Persona de Mediana Edad , Admisión del Paciente , Recuento de Plaquetas , Valor Predictivo de las Pruebas , Pronóstico , Estudios Prospectivos , Factores de Riesgo , Índice de Severidad de la Enfermedad , Trombosis/sangre , Trombosis/diagnóstico , Factores de Tiempo
17.
Platelets ; 32(2): 284-287, 2021 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-33349108

RESUMEN

The frequent finding of thrombocytopenia in patients with severe SARS-CoV-2 infection (COVID-19) and previous evidence that several viruses enter platelets suggest that SARS-CoV-2 might be internalized by platelets of COVID-19. Aim of our study was to assess the presence of SARS-CoV-2 RNA in platelets from hospitalized patients with aconfirmed diagnosis of COVID-19. RNA was extracted from platelets, leukocytes and serum from 24 COVID-19 patients and 3 healthy controls, real-time PCR and ddPCR for viral genes were carried out. SARS-CoV-2 RNA was not detected in any of the samples analyzed nor in healthy controls, by either RT-PCR or ddPCR, while RNA samples from nasopharyngeal swabs of COVID-19 patients were correctly identified. Viral RNA was not detected independently of viral load, of positive nasopharyngeal swabs, or viremia, the last detected in only one patient (4.1%). SARS-CoV-2 entry in platelets is not acommon phenomenon in COVID-19 patients, differently from other viral infections.


Asunto(s)
Plaquetas/virología , COVID-19/sangre , COVID-19/virología , ARN Viral , SARS-CoV-2/fisiología , Anciano , COVID-19/diagnóstico , Prueba de COVID-19 , Femenino , Humanos , Masculino , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa , SARS-CoV-2/aislamiento & purificación , Carga Viral
18.
Platelets ; 32(3): 325-330, 2021 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-33615982

RESUMEN

Platelets play an essential role in maintaining vascular integrity after injury. In addition, platelets contribute to the immune response to pathogens. For instance, they express receptors that mediate binding of viruses, and toll-like receptors that activate the cell in response to pathogen-associated molecular patterns. Platelets can be beneficial and/or detrimental during viral infections. They reduce blood-borne viruses by engulfing the free virus and presenting the virus to neutrophils. However, platelets can also enhance inflammation and tissue injury during viral infections. Here, we discuss the roles of platelets in viral infection.


Asunto(s)
Plaquetas/inmunología , COVID-19/inmunología , Interacciones Huésped-Patógeno/inmunología , Neutrófilos/inmunología , Receptores Virales/inmunología , Proteínas Virales/inmunología , Virus/inmunología , Animales , Plaquetas/patología , Plaquetas/virología , COVID-19/genética , COVID-19/patología , COVID-19/virología , Comunicación Celular/genética , Comunicación Celular/inmunología , Células Dendríticas/inmunología , Células Dendríticas/patología , Células Dendríticas/virología , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Humanos , Inmunidad Innata , Linfocitos/inmunología , Linfocitos/patología , Linfocitos/virología , Neutrófilos/patología , Neutrófilos/virología , Activación Plaquetaria/inmunología , Unión Proteica , Receptores Virales/genética , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Proteínas Virales/genética , Virus/patogenicidad
19.
Int J Mol Sci ; 22(24)2021 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-34948438

RESUMEN

COVID-19 infection is associated with a broad spectrum of presentations, but alveolar capillary microthrombi have been described as a common finding in COVID-19 patients, appearing as a consequence of a severe endothelial injury with endothelial cell membrane disruption. These observations clearly point to the identification of a COVID-19-associated coagulopathy, which may contribute to thrombosis, multi-organ damage, and cause of severity and fatality. One significant finding that emerges in prothrombotic abnormalities observed in COVID-19 patients is that the coagulation alterations are mainly mediated by the activation of platelets and intrinsically related to viral-mediated endothelial inflammation. Beyond the well-known role in hemostasis, the ability of platelets to also release various potent cytokines and chemokines has elevated these small cells from simple cell fragments to crucial modulators in the blood, including their inflammatory functions, that have a large influence on the immune response during infectious disease. Indeed, platelets are involved in the pathogenesis of acute lung injury also by promoting NET formation and affecting vascular permeability. Specifically, the deposition by activated platelets of the chemokine platelet factor 4 at sites of inflammation promotes adhesion of neutrophils on endothelial cells and thrombogenesis, and it seems deeply involved in the phenomenon of vaccine-induced thrombocytopenia and thrombosis. Importantly, the hyperactivated platelet phenotype along with evidence of cytokine storm, high levels of P-selectin, D-dimer, and, on the other hand, decreased levels of fibrinogen, von Willebrand factor, and thrombocytopenia may be considered suitable biomarkers that distinguish the late stage of COVID-19 progression in critically ill patients.


Asunto(s)
Plaquetas/fisiología , COVID-19/sangre , Trombosis/patología , Coagulación Sanguínea , Trastornos de la Coagulación Sanguínea/etiología , Plaquetas/metabolismo , Plaquetas/virología , COVID-19/metabolismo , Síndrome de Liberación de Citoquinas , Células Endoteliales/patología , Productos de Degradación de Fibrina-Fibrinógeno , Hemostasis , Humanos , Inflamación , Fenotipo , Activación Plaquetaria/fisiología , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidad , Trombocitopenia/metabolismo , Trombosis/metabolismo , Trombosis/virología
20.
J Mol Cell Cardiol ; 146: 32-40, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32681845

RESUMEN

SARS-CoV-2 causes a phenotype of pneumonia with diverse manifestation, which is termed as coronavirus disease 2019 (COVID-19). An impressive high transmission rate allows COVID-19 conferring enormous challenge for clinicians worldwide, and developing to a pandemic level. Combined with a series of complications, a part of COVID-19 patients progress into severe cases, which critically contributes to the risk of fatality. To date, coagulopathy has been found as a prominent feature of COVID-19 and severe coagulation dysfunction may be associated with poor prognosis. Coagulopathy in COVID-19 may predispose patients to hypercoagulability-related disorders including thrombosis and even fatal vascular events. Inflammatory storm, uncontrolled inflammation-mediated endothelial injury and renin angiotensin system (RAS) dysregulation are the potential mechanisms. Ongoing efforts made to develop promising therapies provide several potential strategies for hypercoagulability in COVID-19. In this review, we introduce the clinical features of coagulation and the increased vascular thrombotic risk conferred by coagulopathy according to present reports about COVID-19. The potential underlying mechanisms and emerging therapeutic avenues are discussed, emphasizing an urgent need for effective interventions.


Asunto(s)
Anticoagulantes/uso terapéutico , Betacoronavirus/patogenicidad , Infecciones por Coronavirus/complicaciones , Síndrome de Liberación de Citoquinas/complicaciones , Coagulación Intravascular Diseminada/complicaciones , Neumonía Viral/complicaciones , Embolia Pulmonar/complicaciones , Insuficiencia Respiratoria/complicaciones , Enfermedad Aguda , Betacoronavirus/inmunología , Biomarcadores/sangre , Plaquetas/efectos de los fármacos , Plaquetas/patología , Plaquetas/virología , COVID-19 , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/mortalidad , Infecciones por Coronavirus/virología , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Síndrome de Liberación de Citoquinas/mortalidad , Síndrome de Liberación de Citoquinas/virología , Coagulación Intravascular Diseminada/tratamiento farmacológico , Coagulación Intravascular Diseminada/mortalidad , Coagulación Intravascular Diseminada/virología , Productos de Degradación de Fibrina-Fibrinógeno/metabolismo , Heparina/uso terapéutico , Interacciones Huésped-Patógeno/inmunología , Humanos , Pulmón/irrigación sanguínea , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/virología , Pandemias , Tiempo de Tromboplastina Parcial , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/mortalidad , Neumonía Viral/virología , Embolia Pulmonar/tratamiento farmacológico , Embolia Pulmonar/mortalidad , Embolia Pulmonar/virología , Insuficiencia Respiratoria/tratamiento farmacológico , Insuficiencia Respiratoria/mortalidad , Insuficiencia Respiratoria/virología , SARS-CoV-2 , Análisis de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA