Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 339
Filtrar
1.
PLoS Pathog ; 20(6): e1011777, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38913740

RESUMO

COVID-associated coagulopathy seemly plays a key role in post-acute sequelae of SARS- CoV-2 infection. However, the underlying pathophysiological mechanisms are poorly understood, largely due to the lack of suitable animal models that recapitulate key clinical and pathological symptoms. Here, we fully characterized AC70 line of human ACE2 transgenic (AC70 hACE2 Tg) mice for SARS-CoV-2 infection. We noted that this model is highly permissive to SARS-CoV-2 with values of 50% lethal dose and infectious dose as ~ 3 and ~ 0.5 TCID50 of SARS-CoV-2, respectively. Mice infected with 105 TCID50 of SARS-CoV-2 rapidly succumbed to infection with 100% mortality within 5 days. Lung and brain were the prime tissues harboring high viral titers, accompanied by histopathology. However, viral RNA and inflammatory mediators could be detectable in other organs, suggesting the nature of a systemic infection. Lethal challenge of AC70 hACE2 Tg mice caused acute onset of leukopenia, lymphopenia, along with an increased neutrophil-to-lymphocyte ratio (NLR). Importantly, infected animals recapitulated key features of COVID-19-associated coagulopathy. SARS-CoV-2 could induce the release of circulating neutrophil extracellular traps (NETs), along with activated platelet/endothelium marker. Immunohistochemical staining with anti-platelet factor-4 (PF4) antibody revealed profound platelet aggregates especially within blocked veins of the lungs. We showed that acute SARS-CoV-2 infection triggered a hypercoagulable state coexisting with ill-regulated fibrinolysis. Finally, we highlighted the potential role of Annexin A2 (ANXA2) in fibrinolytic failure. ANXA2 is a calcium-dependent phospholipid-binding protein that forms a heterotertrameric complexes localized at the extracellular membranes with two S100A10 small molecules acting as a co-receptor for tissue-plasminogen activator (t-PA), tightly involved in cell surface fibrinolysis. Thus, our results revealing elevated IgG type anti-ANXA2 antibody production, downregulated de novo ANXA2/S100A10 synthesis, and reduced ANXA2/S100A10 association in infected mice, this protein might serve as druggable targets for development of antithrombotic and/or anti-fibrinolytic agents to attenuate pathogenesis of COVID-19.


Assuntos
Enzima de Conversão de Angiotensina 2 , COVID-19 , Modelos Animais de Doenças , Camundongos Transgênicos , SARS-CoV-2 , Animais , COVID-19/patologia , COVID-19/complicações , COVID-19/virologia , COVID-19/metabolismo , Camundongos , Enzima de Conversão de Angiotensina 2/metabolismo , Enzima de Conversão de Angiotensina 2/genética , Humanos , Transtornos da Coagulação Sanguínea/virologia , Transtornos da Coagulação Sanguínea/patologia , Pneumonia Viral/virologia , Pneumonia Viral/patologia , Pneumonia Viral/metabolismo , Betacoronavirus , Pulmão/virologia , Pulmão/patologia , Pulmão/metabolismo , Infecções por Coronavirus/virologia , Infecções por Coronavirus/patologia , Infecções por Coronavirus/complicações , Pandemias , Armadilhas Extracelulares/metabolismo
2.
Clin Exp Pharmacol Physiol ; 49(4): 483-491, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35066912

RESUMO

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.


Assuntos
Lesão Pulmonar Aguda/etiologia , Transtornos da Coagulação Sanguínea/etiologia , COVID-19/patologia , Síndrome da Liberação de Citocina/etiologia , Hemorragia/etiologia , Lipopolissacarídeos/efeitos adversos , Pneumopatias/etiologia , Receptor Tipo 1 de Angiotensina/metabolismo , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Lesão Pulmonar Aguda/patologia , Animais , Transtornos da Coagulação Sanguínea/patologia , COVID-19/etiologia , Síndrome da Liberação de Citocina/patologia , Modelos Animais de Doenças , Hemorragia/patologia , Janus Quinases , Pneumopatias/patologia , Masculino , Ratos , Ratos Wistar
3.
EBioMedicine ; 75: 103812, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35033854

RESUMO

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.


Assuntos
Transtornos da Coagulação Sanguínea/metabolismo , COVID-19/metabolismo , Regulação para Baixo , Endotélio Vascular/metabolismo , Hipóxia/metabolismo , Pulmão/metabolismo , SARS-CoV-2/metabolismo , Trombomodulina/biossíntese , Idoso , Idoso de 80 Anos ou mais , Transtornos da Coagulação Sanguínea/patologia , COVID-19/patologia , Células Endoteliais/metabolismo , Células Endoteliais/ultraestrutura , Endotélio Vascular/ultraestrutura , Feminino , Humanos , Hipóxia/patologia , Pulmão/ultraestrutura , Masculino , Pessoa de Meia-Idade
4.
Acta Pharmacol Sin ; 43(3): 520-528, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34040166

RESUMO

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.


Assuntos
Proteína HMGB1/metabolismo , Insuficiência de Múltiplos Órgãos/patologia , Sepse/patologia , Autofagia/fisiologia , Transtornos da Coagulação Sanguínea/patologia , Síndrome da Liberação de Citocina/patologia , Estresse do Retículo Endoplasmático/fisiologia , Humanos , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Mitocôndrias/patologia , Insuficiência de Múltiplos Órgãos/tratamento farmacológico , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Sepse/tratamento farmacológico , Transdução de Sinais/fisiologia , Receptores Toll-Like/metabolismo
6.
Eur Rev Med Pharmacol Sci ; 25(19): 5904-5912, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34661248

RESUMO

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.


Assuntos
Transtornos da Coagulação Sanguínea/patologia , COVID-19/patologia , Hepatopatias/patologia , Fígado/patologia , Trombose/patologia , Idoso , Autopsia , Biópsia , Eritrócitos/patologia , Fibrina , Hepatócitos/patologia , Humanos , Masculino , Microscopia Eletrônica de Varredura , Pessoa de Meia-Idade , Trombose/complicações , Adulto Jovem
7.
Hamostaseologie ; 41(5): 387-396, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34695855

RESUMO

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.


Assuntos
COVID-19/complicações , SARS-CoV-2 , Pele/irrigação sanguínea , Enzima de Conversão de Angiotensina 2/fisiologia , Anticorpos Antifosfolipídeos/sangue , Transtornos da Coagulação Sanguínea/sangue , Transtornos da Coagulação Sanguínea/etiologia , Transtornos da Coagulação Sanguínea/patologia , COVID-19/patologia , COVID-19/fisiopatologia , Ativação do Complemento , Citocinas/metabolismo , Interações entre Hospedeiro e Microrganismos/imunologia , Interações entre Hospedeiro e Microrganismos/fisiologia , Humanos , Microvasos/imunologia , Microvasos/patologia , Microvasos/fisiopatologia , Pandemias , SARS-CoV-2/patogenicidade , SARS-CoV-2/fisiologia , Pele/imunologia , Vasculite/etiologia , Vasculite/patologia , Vasculite/fisiopatologia , Internalização do Vírus
8.
Clin Immunol ; 232: 108852, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34520860

RESUMO

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.


Assuntos
Coagulação Sanguínea/fisiologia , COVID-19/complicações , Fibrinólise/fisiologia , Inflamação/etiologia , Inflamação/virologia , Adulto , Anticoagulantes/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Transtornos da Coagulação Sanguínea/etiologia , Transtornos da Coagulação Sanguínea/metabolismo , Transtornos da Coagulação Sanguínea/patologia , Transtornos da Coagulação Sanguínea/virologia , COVID-19/metabolismo , COVID-19/patologia , China , Progressão da Doença , Coagulação Intravascular Disseminada/etiologia , Coagulação Intravascular Disseminada/metabolismo , Coagulação Intravascular Disseminada/patologia , Coagulação Intravascular Disseminada/virologia , Feminino , Produtos de Degradação da Fibrina e do Fibrinogênio/metabolismo , Fibrinogênio/metabolismo , Hemorragia/etiologia , Hemorragia/patologia , Hemorragia/virologia , Humanos , Inflamação/patologia , Masculino , Pessoa de Meia-Idade , Tempo de Protrombina , SARS-CoV-2/patogenicidade
9.
Curr Opin Hematol ; 28(6): 445-453, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34232139

RESUMO

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.


Assuntos
Transtornos da Coagulação Sanguínea/patologia , COVID-19/complicações , SARS-CoV-2/isolamento & purificação , Trombose/patologia , Transtornos da Coagulação Sanguínea/epidemiologia , Transtornos da Coagulação Sanguínea/virologia , COVID-19/transmissão , COVID-19/virologia , Humanos , Prognóstico , Trombose/epidemiologia , Trombose/virologia
10.
Sci Rep ; 11(1): 13325, 2021 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-34172793

RESUMO

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.


Assuntos
Anticoagulantes/uso terapêutico , Transtornos da Coagulação Sanguínea , Tratamento Farmacológico da COVID-19 , COVID-19 , Tromboelastografia , Tromboembolia , Trombose Venosa , Idoso , Coagulação Sanguínea , Transtornos da Coagulação Sanguínea/tratamento farmacológico , Transtornos da Coagulação Sanguínea/patologia , COVID-19/sangue , COVID-19/patologia , Estudos de Coortes , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Índice de Gravidade de Doença , Tromboembolia/tratamento farmacológico , Tromboembolia/patologia , Trombose Venosa/tratamento farmacológico , Trombose Venosa/patologia
11.
Diabetes Metab Syndr ; 15(3): 863-868, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33878674

RESUMO

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.


Assuntos
Injúria Renal Aguda/sangue , COVID-19/sangue , Lipídeos/sangue , Injúria Renal Aguda/complicações , Injúria Renal Aguda/epidemiologia , Injúria Renal Aguda/patologia , Adulto , Idoso , Biomarcadores/sangue , Transtornos da Coagulação Sanguínea/sangue , Transtornos da Coagulação Sanguínea/complicações , Transtornos da Coagulação Sanguínea/epidemiologia , Transtornos da Coagulação Sanguínea/patologia , COVID-19/complicações , COVID-19/epidemiologia , COVID-19/patologia , Estudos de Casos e Controles , Estudos de Coortes , Feminino , Humanos , Inflamação/sangue , Inflamação/complicações , Inflamação/epidemiologia , Inflamação/patologia , Masculino , Metaboloma , Pessoa de Meia-Idade , Ohio/epidemiologia , Estudos Prospectivos , SARS-CoV-2/fisiologia , Índice de Gravidade de Doença , Trombose/sangue , Trombose/complicações , Trombose/epidemiologia , Trombose/patologia , Estados Unidos/epidemiologia
12.
A A Pract ; 15(4): e01432, 2021 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-33783367

RESUMO

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.


Assuntos
Lesão Pulmonar Aguda/complicações , Transtornos da Coagulação Sanguínea/complicações , Transtornos da Coagulação Sanguínea/patologia , COVID-19/complicações , COVID-19/patologia , Leucemia Mieloide Aguda/complicações , Lesão Pulmonar Aguda/diagnóstico , Lesão Pulmonar Aguda/patologia , Transtornos da Coagulação Sanguínea/diagnóstico , Lavagem Broncoalveolar , COVID-19/diagnóstico , Citometria de Fluxo , Humanos , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/patologia , Masculino , Pessoa de Meia-Idade , Neutropenia/complicações , Neutropenia/diagnóstico , Neutropenia/patologia , SARS-CoV-2 , Tromboelastografia , Fatores de Virulência
13.
Bull Math Biol ; 83(5): 50, 2021 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-33772645

RESUMO

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.


Assuntos
Coagulação Sanguínea , Modelos Biológicos , Modelagem Computacional Específica para o Paciente , Transtornos da Coagulação Sanguínea/diagnóstico , Transtornos da Coagulação Sanguínea/patologia , Humanos
14.
Am J Respir Cell Mol Biol ; 64(6): 687-697, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33740387

RESUMO

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.


Assuntos
Células Epiteliais Alveolares/metabolismo , Transtornos da Coagulação Sanguínea/metabolismo , COVID-19/metabolismo , Regulação da Expressão Gênica , SARS-CoV-2/metabolismo , Transdução de Sinais , Transcrição Gênica , Células Epiteliais Alveolares/patologia , Células Epiteliais Alveolares/virologia , Transtornos da Coagulação Sanguínea/etiologia , Transtornos da Coagulação Sanguínea/patologia , COVID-19/complicações , COVID-19/patologia , Linhagem Celular , Feminino , Humanos , Vírus da Influenza A/metabolismo , Influenza Humana/complicações , Influenza Humana/metabolismo , Influenza Humana/patologia , Masculino
17.
Korean J Anesthesiol ; 74(4): 350-354, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-32773727

RESUMO

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.


Assuntos
Transtornos da Coagulação Sanguínea/complicações , Transtornos da Coagulação Sanguínea/diagnóstico , COVID-19/complicações , Tromboelastografia/métodos , Trombofilia/complicações , Trombofilia/diagnóstico , Idoso , Transtornos da Coagulação Sanguínea/patologia , Evolução Fatal , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , SARS-CoV-2 , Trombofilia/patologia
19.
Acta Haematol ; 144(2): 166-175, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32506056

RESUMO

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.


Assuntos
Transtornos da Coagulação Sanguínea/psicologia , Qualidade de Vida , Trombofilia/psicologia , Adulto , Idoso , Transtornos da Coagulação Sanguínea/patologia , Estudos Transversais , Feminino , Inquéritos Epidemiológicos , Humanos , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Psicometria , Trombofilia/patologia
20.
Shock ; 55(6): 700-716, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33378321

RESUMO

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.


Assuntos
Transtornos da Coagulação Sanguínea , Coagulação Sanguínea/imunologia , COVID-19 , Imunidade Inata , SARS-CoV-2/imunologia , Transtornos da Coagulação Sanguínea/etiologia , Transtornos da Coagulação Sanguínea/imunologia , Transtornos da Coagulação Sanguínea/patologia , Transtornos da Coagulação Sanguínea/terapia , COVID-19/complicações , COVID-19/patologia , COVID-19/terapia , Humanos , AVC Isquêmico/complicações , AVC Isquêmico/metabolismo , AVC Isquêmico/patologia , Embolia Pulmonar/etiologia , Embolia Pulmonar/imunologia , Embolia Pulmonar/patologia , Embolia Pulmonar/terapia , Trombose/etiologia , Trombose/imunologia , Trombose/patologia , Trombose/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA