Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 340
Filtrar
1.
EBioMedicine ; 75: 103812, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35033854

RESUMEN

BACKGROUND: Thromboembolism is a life-threatening manifestation of coronavirus disease 2019 (COVID-19). We investigated a dysfunctional phenotype of vascular endothelial cells in the lungs during COVID-19. METHODS: We obtained the lung specimens from the patients who died of COVID-19. The phenotype of endothelial cells and immune cells was examined by flow cytometry and immunohistochemistry (IHC) analysis. We tested the presence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in the endothelium using IHC and electron microscopy. FINDINGS: The autopsy lungs of COVID-19 patients exhibited severe coagulation abnormalities, immune cell infiltration, and platelet activation. Pulmonary endothelial cells of COVID-19 patients showed increased expression of procoagulant von Willebrand factor (VWF) and decreased expression of anticoagulants thrombomodulin and endothelial protein C receptor (EPCR). In the autopsy lungs of COVID-19 patients, the number of macrophages, monocytes, and T cells was increased, showing an activated phenotype. Despite increased immune cells, adhesion molecules such as ICAM-1, VCAM-1, E-selectin, and P-selectin were downregulated in pulmonary endothelial cells of COVID-19 patients. Notably, decreased thrombomodulin expression in endothelial cells was associated with increased immune cell infiltration in the COVID-19 patient lungs. There were no SARS-CoV-2 particles detected in the lung endothelium of COVID-19 patients despite their dysfunctional phenotype. Meanwhile, the autopsy lungs of COVID-19 patients showed SARS-CoV-2 virions in damaged alveolar epithelium and evidence of hypoxic injury. INTERPRETATION: Pulmonary endothelial cells become dysfunctional during COVID-19, showing a loss of thrombomodulin expression related to severe thrombosis and infiltration, and endothelial cell dysfunction might be caused by a pathologic condition in COVID-19 patient lungs rather than a direct infection with SARS-CoV-2. FUNDING: This work was supported by the Johns Hopkins University, the American Heart Association, and the National Institutes of Health.


Asunto(s)
Trastornos de la Coagulación Sanguínea/metabolismo , COVID-19/metabolismo , Regulación hacia Abajo , Endotelio Vascular/metabolismo , Hipoxia/metabolismo , Pulmón/metabolismo , SARS-CoV-2/metabolismo , Trombomodulina/biosíntesis , Anciano , Anciano de 80 o más Años , Trastornos de la Coagulación Sanguínea/patología , COVID-19/patología , Células Endoteliales/metabolismo , Células Endoteliales/ultraestructura , Endotelio Vascular/ultraestructura , Femenino , Humanos , Hipoxia/patología , Pulmón/ultraestructura , Masculino , Persona de Mediana Edad
2.
Clin Exp Pharmacol Physiol ; 49(4): 483-491, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35066912

RESUMEN

Progress in the study of Covid-19 disease in rodents has been hampered by the lack of angiotensin-converting enzyme 2 (ACE2; virus entry route to the target cell) affinities for the virus spike proteins across species. Therefore, we sought to determine whether a modified protocol of lipopolysaccharide (LPS)-induced acute respiratory distress syndrome in rats can mimic both cell signalling pathways as well as severe disease phenotypes of Covid-19 disease. Rats were injected via intratracheal (IT) instillation with either 15 mg/kg of LPS (model group) or saline (control group) before being killed after 3 days. A severe acute respiratory syndrome (SARS)-like effect was observed in the model group as demonstrated by the development of a "cytokine storm" (>2.7 fold increase in blood levels of IL-6, IL-17A, GM-CSF, and TNF-α), high blood ferritin, demonstrable coagulopathy, including elevated D-dimer (approximately 10-fold increase), PAI-1, PT, and APTT (p < 0.0001). In addition, LPS increased the expression of lung angiotensin II type I receptor (AT1R)-JAK-STAT axis (>4 fold increase). Chest imaging revealed bilateral small patchy opacities of the lungs. Severe lung injury was noted by the presence of both, alveolar collapse and haemorrhage, desquamation of epithelial cells in the airway lumen, infiltration of inflammatory cells (CD45+ leukocytes), widespread thickening of the interalveolar septa, and ultrastructural alterations similar to Covid-19. Thus, these findings demonstrate that IT injection of 15 mg/kg LPS into rats, induced an AT1R/JAK/STAT-mediated cytokine storm with resultant pneumonia and coagulopathy that was commensurate with moderate and severe Covid-19 disease noted in humans.


Asunto(s)
Lesión Pulmonar Aguda/etiología , Trastornos de la Coagulación Sanguínea/etiología , COVID-19/patología , Síndrome de Liberación de Citoquinas/etiología , Hemorragia/etiología , Lipopolisacáridos/efectos adversos , Enfermedades Pulmonares/etiología , Receptor de Angiotensina Tipo 1/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Lesión Pulmonar Aguda/patología , Animales , Trastornos de la Coagulación Sanguínea/patología , COVID-19/etiología , Síndrome de Liberación de Citoquinas/patología , Modelos Animales de Enfermedad , Hemorragia/patología , Quinasas Janus , Enfermedades Pulmonares/patología , Masculino , Ratas , Ratas Wistar
3.
Acta Pharmacol Sin ; 43(3): 520-528, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34040166

RESUMEN

High mobility group box 1 (HMGB1) is a ubiquitous nuclear protein that is present in almost all cells and regulates the activity of innate immune responses in both intracellular and extracellular settings. Current evidence suggests that HMGB1 plays a pivotal role in human pathological and pathophysiological processes such as the inflammatory response, immune reactions, cell migration, aging, and cell death. Sepsis is a systemic inflammatory response syndrome (SIRS) that occurs in hosts in response to microbial infections with a proven or suspected infectious etiology and is the leading cause of death in intensive care units worldwide, particularly in the aging population. Dysregulated systemic inflammation is a classic characteristic of sepsis, and suppression of HMGB1 may ameliorate inflammation and improve patient outcomes. Here, we focus on the latest breakthroughs regarding the roles of HMGB1 in sepsis and sepsis-related organ injury, the ways by which HMGB1 are released, and the signaling pathways and therapeutics associated with HMGB1. This review highlights recent advances related to HMGB1: the regulation of HMBG1 might be helpful for both basic research and drug development for the treatment of sepsis and sepsis-related organ injury.


Asunto(s)
Proteína HMGB1/metabolismo , Insuficiencia Multiorgánica/patología , Sepsis/patología , Autofagia/fisiología , Trastornos de la Coagulación Sanguínea/patología , Síndrome de Liberación de Citoquinas/patología , Estrés del Retículo Endoplásmico/fisiología , Humanos , Inflamación/patología , Mediadores de Inflamación/metabolismo , Mitocondrias/patología , Insuficiencia Multiorgánica/tratamiento farmacológico , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Sepsis/tratamiento farmacológico , Transducción de Señal/fisiología , Receptores Toll-Like/metabolismo
5.
Eur Rev Med Pharmacol Sci ; 25(19): 5904-5912, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34661248

RESUMEN

OBJECTIVE: Liver injury has been reported in patients with COVID-19. This condition is characterized by severe outcome and could be related with the ability of SARS-CoV-2 to activate cytotoxic T cells. The purpose of this study is to show the histological and scanning electron microscopy features of liver involvement in COVID-19 to characterize the liver changes caused by the activation of multiple molecular pathways following this infection. PATIENTS AND METHODS: Liver biopsies from 4 patients (3 post-mortems and 1 in vivo) with COVID-19 were analyzed with histology and by scanning electron microscopy. RESULTS: The liver changes showed significant heterogeneity. The first case showed ground glass hepatocytes and scattered fibrin aggregates in the sinusoidal lumen. The second evidenced intra-sinusoidal thrombi. The third was characterized by sinusoidal dilatation, atrophy of hepatocytes, Disse's spaces dilatation and intra-sinusoidal aggregates of fibrin and red blood cells. The fourth case exhibited diffuse fibrin aggregates in the dilated Disse spaces and microthrombi in the sinusoidal lumen. CONCLUSIONS: In COVID-19-related liver injury, a large spectrum of pathological changes was observed. The most peculiar features were very mild inflammation, intra-sinusoidal changes, including sinusoidal dilatation, thrombotic sinusoiditis and diffuse intra-sinusoidal fibrin deposition. These findings suggested that a thrombotic sinusoiditis followed by a local diffuse intra-vascular (intra-sinusoidal) coagulation could be the typical features of the SARS-CoV-2-related liver injury.


Asunto(s)
Trastornos de la Coagulación Sanguínea/patología , COVID-19/patología , Hepatopatías/patología , Hígado/patología , Trombosis/patología , Anciano , Autopsia , Biopsia , Eritrocitos/patología , Fibrina , Hepatocitos/patología , Humanos , Masculino , Microscopía Electrónica de Rastreo , Persona de Mediana Edad , Trombosis/complicaciones , Adulto Joven
6.
Hamostaseologie ; 41(5): 387-396, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34695855

RESUMEN

Hypercoagulability and vascular injury, which characterize morbidity in COVID-19 disease, are frequently observed in the skin. Several pathomechanisms, such as inflammation caused by angiotensin-converting enzyme 2-mediated uptake into endothelial cells or SARS-CoV-2-initiated host immune responses, contribute to microthrombus formation and the appearance of vascular skin lesions. Besides pathophysiologic mechanisms observed in the skin, this review describes the clinical appearance of cutaneous vascular lesions and their association with COVID-19 disease, including acro-ischemia, reticular lesions, and cutaneous small vessel vasculitis. Clinicians need to be aware that skin manifestations may be the only symptom in SARS-CoV-2 infection, and that inflammatory and thrombotic SARS-CoV-2-driven processes observed in multiple organs and tissues appear identically in the skin as well.


Asunto(s)
COVID-19/complicaciones , SARS-CoV-2 , Piel/irrigación sanguínea , Enzima Convertidora de Angiotensina 2/fisiología , Anticuerpos Antifosfolípidos/sangre , Trastornos de la Coagulación Sanguínea/sangre , Trastornos de la Coagulación Sanguínea/etiología , Trastornos de la Coagulación Sanguínea/patología , COVID-19/patología , COVID-19/fisiopatología , Activación de Complemento , Citocinas/metabolismo , Interacciones Microbiota-Huesped/inmunología , Interacciones Microbiota-Huesped/fisiología , Humanos , Microvasos/inmunología , Microvasos/patología , Microvasos/fisiopatología , Pandemias , SARS-CoV-2/patogenicidad , SARS-CoV-2/fisiología , Piel/inmunología , Vasculitis/etiología , Vasculitis/patología , Vasculitis/fisiopatología , Internalización del Virus
7.
Clin Immunol ; 232: 108852, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34520860

RESUMEN

BACKGROUND: The majority of the coronavirus disease 2019 (COVID-19) non-survivors meet the criteria for disseminated intravascular coagulation (DIC). Although timely monitoring of clotting hemorrhagic development during the natural course of COVID-19 is critical for understanding pathogenesis, diagnosis, and treatment of the disease, however, limited data are available on the dynamic processes of inflammation/coagulopathy/fibrinolysis (ICF). METHODS: We monitored the dynamic progression of ICF in patients with moderate COVID-19. Out of 694 COVID-19 inpatients from 10 hospitals in Wenzhou, China, we selected 293 adult patients without comorbidities. These patients were divided into different daily cohorts according to the COVID-19 onset-time. Furthermore, data of 223 COVID-19 patients with comorbidities and 22 critical cases were analyzed. Retrospective data were extracted from electronic medical records. RESULTS: The virus-induced damages to pre-hospitalization patients triggered two ICF fluctuations during the 14-day course of the disease. C-reactive protein (CRP), fibrinogen, and D-dimer levels increased and peaked at day 5 (D) 5 and D9 during the 1st and 2nd fluctuations, respectively. The ICF activities were higher during the 2nd fluctuation. Although 12-day medication returned high CRP concentrations to normal and blocked fibrinogen increase, the D-dimer levels remained high on days 17 ±â€¯2 and 23 ±â€¯2 days of the COVID-19 course. Notably, although the oxygenation index, prothrombin time and activated partial thromboplastin time were within the normal range in critical COVID-19 patients at administration, 86% of these patients had a D-dimer level > 500 µg/L. CONCLUSION: COVID-19 is linked with chronic DIC, which could be responsible for the progression of the disease. Understanding and monitoring ICF progression during COVID-19 can help clinicians in identifying the stage of the disease quickly and accurately and administering suitable treatment.


Asunto(s)
Coagulación Sanguínea/fisiología , COVID-19/complicaciones , Fibrinólisis/fisiología , Inflamación/etiología , Inflamación/virología , Adulto , Anticoagulantes/farmacología , Coagulación Sanguínea/efectos de los fármacos , Trastornos de la Coagulación Sanguínea/etiología , Trastornos de la Coagulación Sanguínea/metabolismo , Trastornos de la Coagulación Sanguínea/patología , Trastornos de la Coagulación Sanguínea/virología , COVID-19/metabolismo , COVID-19/patología , China , Progresión de la Enfermedad , Coagulación Intravascular Diseminada/etiología , Coagulación Intravascular Diseminada/metabolismo , Coagulación Intravascular Diseminada/patología , Coagulación Intravascular Diseminada/virología , Femenino , Productos de Degradación de Fibrina-Fibrinógeno/metabolismo , Fibrinógeno/metabolismo , Hemorragia/etiología , Hemorragia/patología , Hemorragia/virología , Humanos , Inflamación/patología , Masculino , Persona de Mediana Edad , Tiempo de Protrombina , SARS-CoV-2/patogenicidad
8.
Curr Opin Hematol ; 28(6): 445-453, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34232139

RESUMEN

PURPOSE OF REVIEW: Coronavirus disease 2019 (COVID-19) is an infectious disease caused by severe acute respiratory syndrome coronavirus-2. Over the past year, COVID-19 has posed a significant threat to global health. Although the infection is associated with mild symptoms in many patients, a significant proportion of patients develop a prothrombotic state due to a combination of alterations in coagulation and immune cell function. The purpose of this review is to discuss the pathophysiological characteristics of COVID-19 that contribute to the immunothrombosis. RECENT FINDINGS: Endotheliopathy during COVID-19 results in increased multimeric von Willebrand factor release and the potential for increased platelet adhesion to the endothelium. In addition, decreased anticoagulant proteins on the surface of endothelial cells further alters the hemostatic balance. Soluble coagulation markers are also markedly dysregulated, including plasminogen activator inhibitor-1 and tissue factor, leading to COVID-19 induced coagulopathy. Platelet hyperreactivity results in increased platelet-neutrophil and -monocyte aggregates further exacerbating the coagulopathy observed during COVID-19. Finally, the COVID-19-induced cytokine storm primes neutrophils to release neutrophil extracellular traps, which trap platelets and prothrombotic proteins contributing to pulmonary thrombotic complications. SUMMARY: Immunothrombosis significantly contributes to the pathophysiology of COVID-19. Understanding the mechanisms behind COVID-19-induced coagulopathy will lead to future therapies for patients.


Asunto(s)
Trastornos de la Coagulación Sanguínea/patología , COVID-19/complicaciones , SARS-CoV-2/aislamiento & purificación , Trombosis/patología , Trastornos de la Coagulación Sanguínea/epidemiología , Trastornos de la Coagulación Sanguínea/virología , COVID-19/transmisión , COVID-19/virología , Humanos , Pronóstico , Trombosis/epidemiología , Trombosis/virología
9.
Sci Rep ; 11(1): 13325, 2021 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-34172793

RESUMEN

COVID 19 is associated with a hypercoagulable state and frequent thromboembolic complications. For how long this acquired abnormality lasts potentially requiring preventive measures, such as anticoagulation remains to be delineated. We used viscoelastic rotational thrombelastometry (ROTEM) in a single center cohort of 13 critical ill patients and performed follow up examinations three months after discharge from ICU. We found clear signs of a hypercoagulable state due to severe hypofibrinolysis and a high rate of thromboembolic complications during the phase of acute illness. Three month follow up revealed normalization of the initial coagulation abnormality and no evidence of venous thrombosis in all thirteen patients. In our cohort the coagulation profile was completely normalized three months after COVID-19. Based on these findings, discontinuation of anticoagulation can be discussed in patients with complete venous reperfusion.


Asunto(s)
Anticoagulantes/uso terapéutico , Trastornos de la Coagulación Sanguínea , Tratamiento Farmacológico de COVID-19 , COVID-19 , Tromboelastografía , Tromboembolia , Trombosis de la Vena , Anciano , Coagulación Sanguínea , Trastornos de la Coagulación Sanguínea/tratamiento farmacológico , Trastornos de la Coagulación Sanguínea/patología , COVID-19/sangre , COVID-19/patología , Estudios de Cohortes , Femenino , Humanos , Masculino , Persona de Mediana Edad , Índice de Severidad de la Enfermedad , Tromboembolia/tratamiento farmacológico , Tromboembolia/patología , Trombosis de la Vena/tratamiento farmacológico , Trombosis de la Vena/patología
10.
Diabetes Metab Syndr ; 15(3): 863-868, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33878674

RESUMEN

BACKGROUND AND AIMS: Reduction of atherogenic lipoproteins is often the ultimate goal of nutritional interventions, however this is complicated given that hypolipidemia is frequently observed in coronavirus disease 2019 (COVID-19) patients. We aimed to explore the association of hypolipidemia with patient outcomes in terms of immunothrombosis and multiorgan injury, focusing on specialized apolipoproteins apo A1 and apo B. METHODS: Lipid profiles of 50 COVID-19 patients and 30 sick controls presenting to the Emergency Department (ED) were measured in this prospective observational study. The primary outcome was development of severe acute kidney injury (AKI). Need for hospitalization and ICU admission were secondary outcomes. Lipoproteins were analyzed for independent association with serum creatinine (SCr) increase ratio and correlated with a wide panel of biomarkers. RESULTS: COVID-19 cohort had significantly lower apo A1 (p = 0.006), and higher apo B/apo A1 ratio (p = 0.041). Patients developing severe AKI had significantly lower LDL-C (p = 0.021). Apo B/apo A1 was associated with 2.25-fold decrease in serum SCr increase ratio, while LDL-C with a 1.5% decrease. Hypolipidemia correlated with low plasminogen, ADAMTS13 activity/VWF:Ag, and high inflammatory biomarkers (CRP, IL-6, IL-8, IL-10), plasminogen activator inhibitor-1 (PAI-1), ED creatinine, and SCr increase ratio. CONCLUSION: Although favored in dietetics, findings of a low LDL-C in COVID-19 patients should be alarming in light of our observations. Low apo B/apo A1 ratio and LDL-C are predictive of renal deterioration in COVID-19 patients, and low LDL-C in particular may potentially serve to indicate COVID-19 related AKI driven by disrupted fibrinolysis and a secondary thrombotic microangiopathy-like process.


Asunto(s)
Lesión Renal Aguda/sangre , COVID-19/sangre , Lípidos/sangre , Lesión Renal Aguda/complicaciones , Lesión Renal Aguda/epidemiología , Lesión Renal Aguda/patología , Adulto , Anciano , Biomarcadores/sangre , Trastornos de la Coagulación Sanguínea/sangre , Trastornos de la Coagulación Sanguínea/complicaciones , Trastornos de la Coagulación Sanguínea/epidemiología , Trastornos de la Coagulación Sanguínea/patología , COVID-19/complicaciones , COVID-19/epidemiología , COVID-19/patología , Estudios de Casos y Controles , Estudios de Cohortes , Femenino , Humanos , Inflamación/sangre , Inflamación/complicaciones , Inflamación/epidemiología , Inflamación/patología , Masculino , Metaboloma , Persona de Mediana Edad , Ohio/epidemiología , Estudios Prospectivos , SARS-CoV-2/fisiología , Índice de Severidad de la Enfermedad , Trombosis/sangre , Trombosis/complicaciones , Trombosis/epidemiología , Trombosis/patología , Estados Unidos/epidemiología
11.
A A Pract ; 15(4): e01432, 2021 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-33783367

RESUMEN

The role of concurrent illness in coronavirus disease 2019 (COVID-19) is unknown. Patients with leukemia may display altered thromboinflammatory responses. We report a 53-year-old man presenting with acute leukemia and COVID-19 who developed thrombotic complications and acute respiratory distress syndrome. Multiple analyses, including rotational thromboelastometry and flow cytometry on blood and bronchoalveolar lavage, are reported to characterize coagulation and immune profiles. The patient developed chemotherapy-induced neutropenia that may have protected his lungs from granulocyte-driven hyperinflammatory acute lung injury. However, neutropenia also alters viral clearing, potentially enabling ongoing viral propagation. This case depicts a precarious equilibrium between leukemia and COVID-19.


Asunto(s)
Lesión Pulmonar Aguda/complicaciones , Trastornos de la Coagulación Sanguínea/complicaciones , Trastornos de la Coagulación Sanguínea/patología , COVID-19/complicaciones , COVID-19/patología , Leucemia Mieloide Aguda/complicaciones , Lesión Pulmonar Aguda/diagnóstico , Lesión Pulmonar Aguda/patología , Trastornos de la Coagulación Sanguínea/diagnóstico , Lavado Broncoalveolar , COVID-19/diagnóstico , Citometría de Flujo , Humanos , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/patología , Masculino , Persona de Mediana Edad , Neutropenia/complicaciones , Neutropenia/diagnóstico , Neutropenia/patología , SARS-CoV-2 , Tromboelastografía , Factores de Virulencia
12.
Am J Respir Cell Mol Biol ; 64(6): 687-697, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33740387

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has rapidly become a global pandemic. In addition to the acute pulmonary symptoms of coronavirus disease (COVID-19) (the disease associated with SARS-CoV-2 infection), pulmonary and distal coagulopathies have caused morbidity and mortality in many patients. Currently, the molecular pathogenesis underlying COVID-19-associated coagulopathies are unknown. Identifying the molecular basis of how SARS-CoV-2 drives coagulation is essential to mitigating short- and long-term thrombotic risks of sick and recovered patients with COVID-19. We aimed to perform coagulation-focused transcriptome analysis of in vitro infected primary respiratory epithelial cells, patient-derived bronchial alveolar lavage cells, and circulating immune cells during SARS-CoV-2 infection. Our objective was to identify transcription-mediated signaling networks driving coagulopathies associated with COVID-19. We analyzed recently published experimentally and clinically derived bulk or single-cell RNA sequencing datasets of SARS-CoV-2 infection to identify changes in transcriptional regulation of blood coagulation. We also confirmed that the transcriptional expression of a key coagulation regulator was recapitulated at the protein level. We specifically focused our analysis on lung tissue-expressed genes regulating the extrinsic coagulation cascade and the plasminogen activation system. Analyzing transcriptomic data of in vitro infected normal human bronchial epithelial cells and patient-derived bronchial alveolar lavage samples revealed that SARS-CoV-2 infection induces the extrinsic blood coagulation cascade and suppresses the plasminogen activation system. We also performed in vitro SARS-CoV-2 infection experiments on primary human lung epithelial cells to confirm that transcriptional upregulation of tissue factor, the extrinsic coagulation cascade master regulator, manifested at the protein level. Furthermore, infection of normal human bronchial epithelial cells with influenza A virus did not drive key regulators of blood coagulation in a similar manner as SARS-CoV-2. In addition, peripheral blood mononuclear cells did not differentially express genes regulating the extrinsic coagulation cascade or plasminogen activation system during SARS-CoV-2 infection, suggesting that they are not directly inducing coagulopathy through these pathways. The hyperactivation of the extrinsic blood coagulation cascade and the suppression of the plasminogen activation system in SARS-CoV-2-infected epithelial cells may drive diverse coagulopathies in the lung and distal organ systems. Understanding how hosts drive such transcriptional changes with SARS-CoV-2 infection may enable the design of host-directed therapeutic strategies to treat COVID-19 and other coronaviruses inducing hypercoagulation.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Trastornos de la Coagulación Sanguínea/metabolismo , COVID-19/metabolismo , Regulación de la Expresión Génica , SARS-CoV-2/metabolismo , Transducción de Señal , Transcripción Genética , Células Epiteliales Alveolares/patología , Células Epiteliales Alveolares/virología , Trastornos de la Coagulación Sanguínea/etiología , Trastornos de la Coagulación Sanguínea/patología , COVID-19/complicaciones , COVID-19/patología , Línea Celular , Femenino , Humanos , Virus de la Influenza A/metabolismo , Gripe Humana/complicaciones , Gripe Humana/metabolismo , Gripe Humana/patología , Masculino
13.
Bull Math Biol ; 83(5): 50, 2021 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-33772645

RESUMEN

Blood coagulation represents one of the most studied processes in biomedical modelling. However, clinical applications of this modelling remain limited because of the complexity of this process and because of large inter-patient variation of the concentrations of blood factors, kinetic constants and physiological conditions. Determination of some of these patients-specific parameters is experimentally possible, but it would be related to excessive time and material costs impossible in clinical practice. We propose in this work a methodological approach to patient-specific modelling of blood coagulation. It begins with conventional thrombin generation tests allowing the determination of parameters of a reduced kinetic model. Next, this model is used to study spatial distributions of blood factors and blood coagulation in flow, and to evaluate the results of medical treatment of blood coagulation disorders.


Asunto(s)
Coagulación Sanguínea , Modelos Biológicos , Modelación Específica para el Paciente , Trastornos de la Coagulación Sanguínea/diagnóstico , Trastornos de la Coagulación Sanguínea/patología , Humanos
15.
Korean J Anesthesiol ; 74(4): 350-354, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-32773727

RESUMEN

BACKGROUND: Coronavirus disease (COVID-19)-associated coagulopathy is most often characterized by elevated D-dimer, interleukin-6, and plasma fibrinogen concentrations as well as hypercoagulability in thromboelastometry with increased clot firmness in the EXTEM, INTEM, and FIBTEM assays. Clinically, it manifests with a very high incidence of thrombosis, particularly in the pulmonary system, whereas bleeding complications are infrequent. CASE: Here, we describe two critically ill patients with COVID-19 admitted to our intensive care unit demonstrating different thromboelastometry and biomarker patterns. One patient presented with hypercoagulability and the other patient with hypocoagulability and fibrinolysis shutdown in thromboelastometry. The pathophysiology and the potential impact on treatment options are discussed. CONCLUSIONS: A combination of biomarkers and thromboelastometry results can be helpful in the future to decide which therapeutic strategy might be most appropriate for critically ill patients with COVID-19. This would be an important step to establish precision medicine in this high-risk patient population.


Asunto(s)
Trastornos de la Coagulación Sanguínea/complicaciones , Trastornos de la Coagulación Sanguínea/diagnóstico , COVID-19/complicaciones , Tromboelastografía/métodos , Trombofilia/complicaciones , Trombofilia/diagnóstico , Anciano , Trastornos de la Coagulación Sanguínea/patología , Resultado Fatal , Femenino , Humanos , Masculino , Persona de Mediana Edad , SARS-CoV-2 , Trombofilia/patología
17.
Acta Haematol ; 144(2): 166-175, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32506056

RESUMEN

BACKGROUND/AIMS: The newly adapted generic KINDL-A(dult)B(rief) questionnaire showed satisfactory cross-sectional psychometric properties in adults with bleeding disorders or thrombophilia. This investigation aimed to evaluate its cross-sectional and longitudinal construct validity. METHODS: After ethical committee approval and written informed consent, 335 patients (mean age 51.8 ± 16.6 years, 60% women) with either predominant thrombophilia (n = 260) or predominant bleeding disorders (n = 75) participated. At baseline, patients answered the KINDL-AB, the MOS 36-item Short-Form Health Survey (SF-36), and the EQ-5D-3L. A subgroup of 117 patients repeated the questionnaire after a median follow-up of 2.6 years (range: 0.4-3.5). A priori hypotheses were evaluated regarding convergent correlations between KINDL-AB overall well-being and specific subscales, EQ-5D-3L index values (EQ-IV), EQ-5D visual analog scale (EQ-VAS), and SF-36 subscales. RESULTS: Contrary to hypothesis, baseline correlations between the KINDL-AB and EQ-IV/EQ-VAS were all moderate while, as hypothesized, several KINDL-AB subscales and SF-36 subscales correlated strongly. At follow-up, no significant changes in all three instruments occurred. Correlations between instruments over the follow-up were mostly moderate and partially strong. Contrary to hypothesis but consistent with no significant changes in health-related quality of life, convergent correlations between changes in KINDL-AB overall well-being, physical and psychological well-being, and EQ-IV/EQ-VAS were all weak. CONCLUSIONS: While repeated measures of KINDL-AB showed moderate to strong correlations, changes in KINDL-AB overall well-being and subscales correlated more weakly than expected with changes involving two established instruments of generic health status.


Asunto(s)
Trastornos de la Coagulación Sanguínea/psicología , Calidad de Vida , Trombofilia/psicología , Adulto , Anciano , Trastornos de la Coagulación Sanguínea/patología , Estudios Transversales , Femenino , Encuestas Epidemiológicas , Humanos , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Psicometría , Trombofilia/patología
19.
Shock ; 55(6): 700-716, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33378321

RESUMEN

ABSTRACT: There is increasing evidence that novel coronavirus disease 2019 (COVID-19) leads to a significant coagulopathy, a phenomenon termed "COVID-19 associated coagulopathy." COVID-19 has been associated with increased rates of both venous and arterial thromboembolic events, a source of significant morbidity and mortality in this disease. Further evidence suggests a link between the inflammatory response and coagulopathy associated with COVID-19. This presents a unique set of challenges for diagnosis, prevention, and treatment of thrombotic complications. In this review, we summarize and discuss the current literature on laboratory coagulation disruptions associated with COVID-19 and the clinical effects of thromboembolic events including pulmonary embolism, deep vein thrombosis, peripheral arterial thrombosis, and acute ischemic stroke in COVID-19. Endothelial injury and augmented innate immune response are implicated in the development of diffuse macro- and microvascular thrombosis in COVID-19. The pathophysiology of COVID-19 associated coagulopathy is an important determinant of appropriate treatment and monitoring of these complications. We highlight the importance of diagnosis and management of dysregulated coagulation in COVID-19 to improve outcomes in COVID-19 patients with thromboembolic complications.


Asunto(s)
Trastornos de la Coagulación Sanguínea , Coagulación Sanguínea/inmunología , COVID-19 , Inmunidad Innata , SARS-CoV-2/inmunología , Trastornos de la Coagulación Sanguínea/etiología , Trastornos de la Coagulación Sanguínea/inmunología , Trastornos de la Coagulación Sanguínea/patología , Trastornos de la Coagulación Sanguínea/terapia , COVID-19/complicaciones , COVID-19/patología , COVID-19/terapia , Humanos , Accidente Cerebrovascular Isquémico/complicaciones , Accidente Cerebrovascular Isquémico/metabolismo , Accidente Cerebrovascular Isquémico/patología , Embolia Pulmonar/etiología , Embolia Pulmonar/inmunología , Embolia Pulmonar/patología , Embolia Pulmonar/terapia , Trombosis/etiología , Trombosis/inmunología , Trombosis/patología , Trombosis/terapia
20.
Semin Pediatr Surg ; 29(5): 150966, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33069295

RESUMEN

Vascular anomalies, comprised of vascular tumors and malformations, are frequently associated with coagulopathy. Recognition of and familiarity with these vascular anomaly-associated hematologic abnormalities prior to surgery or interventional procedures is essential for pre-operative pre-operative planning. Complicated coagulopathies present within the framework of either Kasabach-Merritt phenomenon (KMP) or localized intravascular coagulopathy (LIC), and their management benefits from the expertise of a hematologist for optimal intra- and peri­operative care. Furthermore, with the recent broadening of understanding of vascular anomalies and the addition of new classification sub-groups, distinctions of these two classic coagulopathy phenotypes have been recognized. This review summarizes the main features of these coagulopathies, described according to their vascular anomaly type, highlighting clinical aspects relevant to surgical management.


Asunto(s)
Trastornos de la Coagulación Sanguínea , Síndrome de Kasabach-Merritt , Malformaciones Vasculares , Neoplasias Vasculares , Trastornos de la Coagulación Sanguínea/diagnóstico , Trastornos de la Coagulación Sanguínea/patología , Trastornos de la Coagulación Sanguínea/cirugía , Niño , Humanos , Síndrome de Kasabach-Merritt/diagnóstico , Síndrome de Kasabach-Merritt/patología , Síndrome de Kasabach-Merritt/cirugía , Malformaciones Vasculares/diagnóstico , Malformaciones Vasculares/patología , Malformaciones Vasculares/cirugía , Neoplasias Vasculares/diagnóstico , Neoplasias Vasculares/patología , Neoplasias Vasculares/cirugía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...