Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 208
Filtrar
1.
Ren Fail ; 46(2): 2374448, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38973428

RESUMO

BACKGROUND: Patients with idiopathic membranous nephropathy (IMN) are more likely to be complicated by venous thromboembolism (VTE). The aim of the study was to investigate the potential association between anti-phospholipase A2 receptor (PLA2R) antibodies and hypercoagulability in patients with IMN. METHODS: A total of 168 patients with biopsy-proven IMN and 36 patients with biopsy-proven minimal change disease (MCD) were enrolled in this study. The clinical data, serum anti-PLA2R antibodies and coagulation-related indices of the patients were retrospectively analyzed. RESULTS: Patients with IMN were categorized into glomerular PLA2R staining-positive (GAg+) IMN group and glomerular PLA2R staining-negative (GAg-) IMN group in the study. Patients with IMN who were GAg + had lower PT, APTT and R time than patients with IMN who were GAg-, while the CI value was higher in patients with IMN who were GAg+. Patients with IMN who were GAg + were divided into the SAb+/GAg + group and the SAb-/GAg + group. Patients with IMN who were SAb+/GAg + had higher Fib and MA values than patients with IMN who were SAb-/GAg+. Correlation analysis showed that serum anti-PLA2R antibodies were positively correlated with fibrinogen, D-dimer, K time, CI value, α-angle, and MA value. Multiple linear regression analysis indicated that anti-PLA2R antibodies were independently correlated with fibrinogen and MA value. CONCLUSION: Our study provides a new perspective on the underlying mechanisms of hypercoagulability in patients with IMN. Anti-PLA2R antibodies are associated with hypercoagulability in patients with IMN and may affect coagulation in patients with IMN by affecting platelet aggregation function and fibrinogen counts.


Assuntos
Autoanticorpos , Glomerulonefrite Membranosa , Receptores da Fosfolipase A2 , Trombofilia , Humanos , Receptores da Fosfolipase A2/imunologia , Glomerulonefrite Membranosa/sangue , Glomerulonefrite Membranosa/imunologia , Glomerulonefrite Membranosa/complicações , Masculino , Feminino , Estudos Retrospectivos , Pessoa de Meia-Idade , Adulto , Trombofilia/etiologia , Trombofilia/imunologia , Trombofilia/sangue , Autoanticorpos/sangue
2.
Medicine (Baltimore) ; 103(29): e38929, 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-39029039

RESUMO

RATIONALE: The phenomenon of hypercoagulability has not been previously documented in individuals with Morvan's syndrome, especially in those associated with contactin-associated protein-like receptor 2 (CASPR2). PATIENT CONCERNS: A previously healthy 32-year-old Chinese male was admitted to the hospital with central and peripheral neurologic symptoms. The patient was tested positive for anti-CASPR2 antibodies, and also presented with an activated coagulation state on admission, characterized by a low activated partial thromboplastin time and a high platelet count. With gradual improvement of clinical symptoms, activated partial thromboplastin time, and platelet count returned to normal. Simultaneously, anti-CASPR2 antibody titers significantly decreased and eventually became undetectable. DIAGNOSES: The patient was diagnosed as Morvan's syndrome with positive anti-CASPAR2 antibodies accompanied with hypercoagulable state. INTERVENTIONS: Plasmapheresis was administered to improve the symptoms combined with prednisolone acetate therapy. OUTCOMES: The patient experienced complete resolution of all symptoms during hospitalization and generally recovery after 2 months of discharge. LESSONS: Emphasis should be directed towards hypercoagulability in individuals diagnosed with Morvan's syndrome, particularly those presenting with positive anti-CASPR2 antibodies. Anticoagulant therapy may represent a novel therapeutic approach for individuals afflicted with Morvan's syndrome and exhibiting positivity for anti-CASPR2 antibodies.


Assuntos
Autoanticorpos , Proteínas de Membrana , Proteínas do Tecido Nervoso , Trombofilia , Humanos , Masculino , Adulto , Trombofilia/imunologia , Trombofilia/tratamento farmacológico , Autoanticorpos/sangue , Autoanticorpos/imunologia , Proteínas de Membrana/imunologia , Proteínas do Tecido Nervoso/imunologia , Plasmaferese
3.
Clin Appl Thromb Hemost ; 27: 10760296211045902, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34590493

RESUMO

INTRODUCTION: Diabetes is the most common of comorbidity in patients with SARS-COV-2 pneumonia. Coagulation abnormalities with D-dimer levels are increased in this disease. OBJECTIFS: We aimed to compare the levels of D-dimer in diabetic and non-diabetic patients with COVID 19. A link between D-dimer and mortality has also been established. MATERIALS: A retrospective study was carried out at the University Hospital Center of Oujda (Morocco) from November 01st to December 01st, 2020. Our study population was divided into two groups: a diabetic group and a second group without diabetes to compare clinical and biological characteristics between the two groups. In addition, the receiver operator characteristic curve was used to assess the optimal D-dimer cut-off point for predicting mortality in diabetics. RESULTS: 201 confirmed-COVID-19-patients were included in the final analysis. The median age was 64 (IQR 56-73), and 56% were male. Our study found that D-dimer levels were statistically higher in diabetic patients compared to non-diabetic patients. (1745 vs 845 respectively, P = 0001). D-dimer level > 2885 ng/mL was a significant predictor of mortality in diabetic patients with a sensitivity of 71,4% and a specificity of 70,7%. CONCLUSION: Our study found that diabetics with COVID-19 are likely to develop hypercoagulation with a poor prognosis.


Assuntos
COVID-19/sangue , Diabetes Mellitus/sangue , Produtos de Degradação da Fibrina e do Fibrinogênio/análise , SARS-CoV-2 , Trombofilia/sangue , Idoso , Área Sob a Curva , Biomarcadores , Proteína C-Reativa/análise , COVID-19/complicações , COVID-19/epidemiologia , Comorbidade , Complicações do Diabetes/sangue , Complicações do Diabetes/epidemiologia , Diabetes Mellitus/epidemiologia , Feminino , Mortalidade Hospitalar , Humanos , Hipertensão/epidemiologia , Inflamação/imunologia , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Estresse Oxidativo , Prognóstico , Curva ROC , Estudos Retrospectivos , Fatores de Risco , Trombofilia/etiologia , Trombofilia/imunologia
4.
Cytokine Growth Factor Rev ; 60: 52-60, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34090785

RESUMO

Antiphospholipid antibodies (aPLs), present in 1-5 % of healthy individuals, are associated with the risk of antiphospholipid syndrome (APS), which is the most common form of acquired thrombophilia. APLs may appear following infections or vaccinations and have been reported in patients with COronaVIrus Disease-2019 (COVID-19). However, their association with COVID-19 vaccination is unclear. Notably, a few cases of thrombocytopenia and thrombotic events resembling APS have been reported to develop in recipients of either adenoviral vector- or mRNA-based COVID-19 vaccines. The aim of this review is therefore to speculate on the plausible role of aPLs in the pathogenesis of these rare adverse events. Adenoviral vector-based vaccines can bind platelets and induce their destruction in the reticuloendothelial organs. Liposomal mRNA-based vaccines may instead favour activation of coagulation factors and confer a pro-thrombotic phenotype to endothelial cells and platelets. Furthermore, both formulations may trigger a type I interferon response associated with the generation of aPLs. In turn, aPLs may lead to aberrant activation of the immune response with participation of innate immune cells, cytokines and the complement cascade. NETosis, monocyte recruitment and cytokine release may further support endothelial dysfunction and promote platelet aggregation. These considerations suggest that aPLs may represent a risk factor for thrombotic events following COVID-19 vaccination, and deserve further investigations.


Assuntos
Anticorpos Antifosfolipídeos/análise , Anticorpos Antifosfolipídeos/imunologia , Vacinas contra COVID-19/efeitos adversos , Vacinas contra COVID-19/imunologia , Trombofilia/etiologia , Síndrome Antifosfolipídica/etiologia , Síndrome Antifosfolipídica/imunologia , Contraindicações de Medicamentos , Humanos , Trombofilia/imunologia
5.
Cells ; 10(4)2021 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-33923537

RESUMO

Coronavirus disease 2019 (COVID-19) is the most devastating infectious disease in the 21st century with more than 2 million lives lost in less than a year. The activation of inflammasome in the host infected by SARS-CoV-2 is highly related to cytokine storm and hypercoagulopathy, which significantly contribute to the poor prognosis of COVID-19 patients. Even though many studies have shown the host defense mechanism induced by inflammasome against various viral infections, mechanistic interactions leading to downstream cellular responses and pathogenesis in COVID-19 remain unclear. The SARS-CoV-2 infection has been associated with numerous cardiovascular disorders including acute myocardial injury, myocarditis, arrhythmias, and venous thromboembolism. The inflammatory response triggered by the activation of NLRP3 inflammasome under certain cardiovascular conditions resulted in hyperinflammation or the modulation of angiotensin-converting enzyme 2 signaling pathways. Perturbations of several target cells and tissues have been described in inflammasome activation, including pneumocytes, macrophages, endothelial cells, and dendritic cells. The interplay between inflammasome activation and hypercoagulopathy in COVID-19 patients is an emerging area to be further addressed. Targeted therapeutics to suppress inflammasome activation may have a positive effect on the reduction of hyperinflammation-induced hypercoagulopathy and cardiovascular disorders occurring as COVID-19 complications.


Assuntos
COVID-19/complicações , Doenças Cardiovasculares/etiologia , Inflamassomos/imunologia , Trombofilia/etiologia , Animais , COVID-19/imunologia , COVID-19/patologia , Doenças Cardiovasculares/imunologia , Doenças Cardiovasculares/patologia , Humanos , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , SARS-CoV-2/imunologia , Trombofilia/imunologia , Trombofilia/patologia
6.
Fertil Steril ; 115(3): 561-566, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33610320

RESUMO

Certain miscarriages result from immunologic factors, but there is no clear identification of the precise causes of recurrent pregnancy loss (RPL). Miscarriages and RPL can arise from a disruption of maternal-fetal immune homeostasis. Remodeling of the maternal uterine spiral arteries is one of the key steps for normal growth and development of the fetus. An adequate oxygen supply is necessary for correct placentation, and it is accomplished by proper vascular changes. The development of fetal tissues creates a potential immunologic problem since the fetus can express paternal antigens and, in some cases, antigens of a gamete donor. The maternal immune system actively responds to fetal antigens, and dysregulation of this crosstalk could partly explain pregnancy complications such as miscarriages and RPL. RPL resulting from thrombophilia is primarily due to acquired thrombophilia, and therefore screening and treatment should be focused on antiphospholipid antibody syndrome.


Assuntos
Aborto Habitual/imunologia , Tolerância Imunológica/imunologia , Fatores Imunológicos/imunologia , Placentação/imunologia , Trombofilia/imunologia , Aborto Habitual/diagnóstico , Aborto Habitual/etiologia , Feminino , Humanos , Gravidez , Trombofilia/complicações , Trombofilia/diagnóstico
8.
Arthritis Rheumatol ; 73(1): 23-35, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32929876

RESUMO

The clinical progression of the severe acute respiratory syndrome coronavirus 2 infection, coronavirus 2019 (COVID-19), to critical illness is associated with an exaggerated immune response, leading to magnified inflammation termed the "cytokine storm." This response is thought to contribute to the pathogenicity of severe COVID-19. There is an initial weak interferon response and macrophage activation that results in delayed neutrophil recruitment leading to impeded viral clearance. This causes prolonged immune stimulation and the release of proinflammatory cytokines. Elevated inflammatory markers in COVID-19 (e.g., d-dimer, C-reactive protein, lactate dehydrogenase, ferritin, and interleukin-6) are reminiscent of the cytokine storm seen in severe hyperinflammatory macrophage disorders. The dysfunctional immune response in COVID-19 also includes lymphopenia, reduced T cells, reduced natural killer cell maturation, and unmitigated plasmablast proliferation causing aberrant IgG levels. The progression to severe disease is accompanied by endotheliopathy, immunothrombosis, and hypercoagulability. Thus, both parts of the immune system-innate and adaptive-play a significant role in the cytokine storm, multiorgan dysfunction, and coagulopathy. This review highlights the importance of understanding the immunologic mechanisms of COVID-19 as they inform the clinical presentation and advise potential therapeutic targets.


Assuntos
Imunidade Adaptativa/imunologia , COVID-19/imunologia , Síndrome da Liberação de Citocina/imunologia , Imunidade Inata/imunologia , Síndrome do Desconforto Respiratório/imunologia , Formação de Anticorpos , Antivirais/uso terapêutico , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , COVID-19/sangue , COVID-19/fisiopatologia , Inativadores do Complemento/uso terapêutico , Citocinas/antagonistas & inibidores , Citocinas/imunologia , Endotélio Vascular/imunologia , Endotélio Vascular/fisiopatologia , Humanos , Imunidade Humoral/imunologia , Imunoglobulina G/imunologia , Fatores Imunológicos/uso terapêutico , Memória Imunológica , Imunossupressores/uso terapêutico , Interferons/imunologia , Células Matadoras Naturais/imunologia , Linfopenia/imunologia , Ativação de Macrófagos/imunologia , Infiltração de Neutrófilos/imunologia , SARS-CoV-2 , Síndrome de Resposta Inflamatória Sistêmica/imunologia , Trombofilia/sangue , Trombofilia/imunologia , Trombose/sangue , Trombose/imunologia , Tratamento Farmacológico da COVID-19
10.
Sci Transl Med ; 12(570)2020 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-33139519

RESUMO

Patients with COVID-19 are at high risk for thrombotic arterial and venous occlusions. Lung histopathology often reveals fibrin-based blockages in the small blood vessels of patients who succumb to the disease. Antiphospholipid syndrome is an acquired and potentially life-threatening thrombophilia in which patients develop pathogenic autoantibodies targeting phospholipids and phospholipid-binding proteins (aPL antibodies). Case series have recently detected aPL antibodies in patients with COVID-19. Here, we measured eight types of aPL antibodies in serum samples from 172 patients hospitalized with COVID-19. These aPL antibodies included anticardiolipin IgG, IgM, and IgA; anti-ß2 glycoprotein I IgG, IgM, and IgA; and anti-phosphatidylserine/prothrombin (aPS/PT) IgG and IgM. We detected aPS/PT IgG in 24% of serum samples, anticardiolipin IgM in 23% of samples, and aPS/PT IgM in 18% of samples. Antiphospholipid autoantibodies were present in 52% of serum samples using the manufacturer's threshold and in 30% using a more stringent cutoff (≥40 ELISA-specific units). Higher titers of aPL antibodies were associated with neutrophil hyperactivity, including the release of neutrophil extracellular traps (NETs), higher platelet counts, more severe respiratory disease, and lower clinical estimated glomerular filtration rate. Similar to IgG from patients with antiphospholipid syndrome, IgG fractions isolated from patients with COVID-19 promoted NET release from neutrophils isolated from healthy individuals. Furthermore, injection of IgG purified from COVID-19 patient serum into mice accelerated venous thrombosis in two mouse models. These findings suggest that half of patients hospitalized with COVID-19 become at least transiently positive for aPL antibodies and that these autoantibodies are potentially pathogenic.


Assuntos
Anticorpos Antifosfolipídeos/sangue , COVID-19/imunologia , Trombose Venosa/etiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Anticorpos Antifosfolipídeos/administração & dosagem , Síndrome Antifosfolipídica/sangue , Síndrome Antifosfolipídica/etiologia , Síndrome Antifosfolipídica/imunologia , COVID-19/sangue , COVID-19/complicações , Estudos de Coortes , Estudos Transversais , Modelos Animais de Doenças , Armadilhas Extracelulares/metabolismo , Feminino , Humanos , Imunoglobulina G/administração & dosagem , Imunoglobulina G/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Neutrófilos/imunologia , Neutrófilos/metabolismo , Trombofilia/sangue , Trombofilia/etiologia , Trombofilia/imunologia , Pesquisa Translacional Biomédica , Trombose Venosa/sangue , Trombose Venosa/imunologia
11.
Pediatr Blood Cancer ; 67(12): e28745, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33009893

RESUMO

Infection from severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), though mainly a respiratory disease, can impair many systems, including causing hematological complications. Lymphopenia and hypercoagulability have been reported in adults with coronavirus disease 2019 (COVID-19) and are considered markers of poor prognosis. This review summarizes the hematological findings in children with SARS-CoV-2 infection. The majority of infected children had a normal leukocyte count, while the most common white blood cell abnormality was leukopenia. Lymphopenia, which may be a marker of severe disease, was rarer in children than in adults, possibly due to their immature immune system or due to the less severe manifestation of COVID-19 in this age group. Age may have an impact, and in neonates and infants the most common abnormality was lymphocytosis. Abnormalities of red blood cells and platelets were uncommon. Anemia and hypercoagulability were reported mainly in children presenting the novel multisystem inflammatory syndrome (MIS) associated with SARS-CoV-2.


Assuntos
Anemia/sangue , Betacoronavirus/metabolismo , Infecções por Coronavirus/sangue , Linfopenia/sangue , Pandemias , Pneumonia Viral/sangue , Trombofilia/sangue , Adolescente , Anemia/epidemiologia , Anemia/imunologia , Betacoronavirus/imunologia , Biomarcadores/sangue , Plaquetas/imunologia , Plaquetas/metabolismo , COVID-19 , Criança , Pré-Escolar , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/imunologia , Eritrócitos/imunologia , Eritrócitos/metabolismo , Feminino , Humanos , Lactente , Recém-Nascido , Contagem de Leucócitos , Linfopenia/epidemiologia , Linfopenia/imunologia , Masculino , Pneumonia Viral/epidemiologia , Pneumonia Viral/imunologia , SARS-CoV-2 , Trombofilia/epidemiologia , Trombofilia/imunologia
12.
Transfusion ; 60 Suppl 6: S101-S121, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33089936

RESUMO

Viscoelastic tests (VETs) have been used routinely for liver transplantation, cardiac surgery, and trauma, but only recently have found clinical utility in benign hematologic disorders. Therefore, guidelines for diagnosis and treatment of these disorders based on viscoelastic variables have been adapted from the existing transplant, cardiothoracic surgery, and trauma resuscitation literature. As a result, diagnostic and therapeutic strategies for benign hematologic disorders utilizing VETs are not uniform. Accordingly, even though there has been a recent increase in the utilization of VET for the diagnosis and treatment of such disorders, the literature is still in its early stages. Analysis of point-of-care viscoelastic tracings from benign hematologic disorders has the potential to allow prompt recognition of disease and to guide patient-specific intervention. Here we present a review describing the application of VETs to benign hematologic disorders.


Assuntos
Doenças Hematológicas/sangue , Testes Imediatos , Tromboelastografia , Animais , Doenças Autoimunes/sangue , Transtornos da Coagulação Sanguínea/sangue , Transtornos da Coagulação Sanguínea/genética , Transfusão de Componentes Sanguíneos , Modelos Animais de Doenças , Previsões , Hemostasia/fisiologia , Humanos , Tromboelastografia/instrumentação , Tromboelastografia/métodos , Trombofilia/sangue , Trombofilia/etiologia , Trombofilia/genética , Trombofilia/imunologia , Vasculite/sangue
15.
Aging (Albany NY) ; 12(16): 15954-15961, 2020 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-32826388

RESUMO

The COVID-19 pandemic has caused monumental mortality, and there are still no adequate therapies. Most severely ill COVID-19 patients manifest a hyperactivated immune response, instigated by interleukin 6 (IL6) that triggers a so called "cytokine storm" and coagulopathy. Hypoxia is also associated with COVID-19. So far overlooked is the fact that both IL6 and hypoxia depress the abundance of a key anticoagulant, Protein S. We speculate that the IL6-driven cytokine explosion plus hypoxemia causes a severe drop in Protein S level that exacerbates the thrombotic risk in COVID-19 patients. Here we highlight a mechanism by which the IL6-hypoxia curse causes a deadly hypercoagulable state in COVID-19 patients, and we suggest a path to therapy.


Assuntos
Infecções por Coronavirus , Síndrome da Liberação de Citocina , Hipóxia , Pandemias , Pneumonia Viral , Proteína S , Trombofilia/imunologia , Enzima de Conversão de Angiotensina 2 , Anticoagulantes/metabolismo , Anticoagulantes/farmacologia , Betacoronavirus/fisiologia , COVID-19 , Infecções por Coronavirus/sangue , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/terapia , Síndrome da Liberação de Citocina/sangue , Síndrome da Liberação de Citocina/virologia , Gerenciamento Clínico , Humanos , Hipóxia/sangue , Hipóxia/etiologia , Hipóxia/imunologia , Interleucina-6/sangue , Peptidil Dipeptidase A/metabolismo , Pneumonia Viral/sangue , Pneumonia Viral/imunologia , Pneumonia Viral/terapia , Proteína S/metabolismo , Proteína S/farmacologia , SARS-CoV-2 , Índice de Gravidade de Doença
16.
Am J Hematol ; 95(12): 1578-1589, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32857878

RESUMO

Thrombotic complications are frequent in COVID-19 and contribute significantly to mortality and morbidity. We review several mechanisms of hypercoagulability in sepsis that may be upregulated in COVID-19. These include immune-mediated thrombotic mechanisms, complement activation, macrophage activation syndrome, antiphospholipid antibody syndrome, hyperferritinemia, and renin-angiotensin system dysregulation. We highlight biomarkers within each pathway with potential prognostic value in COVID-19. Lastly, recent observational studies have evaluated a role for the expanded use of therapeutic anticoagulation in COVID-19. We review strengths and weaknesses of these studies, and we also discuss the hypothetical benefit and anticipated challenges of fibrinolytic therapy in COVID-19.


Assuntos
COVID-19/complicações , SARS-CoV-2 , Trombose/epidemiologia , Anticoagulantes/uso terapêutico , Síndrome Antifosfolipídica , COVID-19/imunologia , COVID-19/terapia , Ativação do Complemento , Estado Terminal/epidemiologia , Síndrome da Liberação de Citocina/epidemiologia , Coagulação Intravascular Disseminada , Ferritinas/sangue , Humanos , Hiperferritinemia/epidemiologia , Ativação de Macrófagos , Embolia Pulmonar/epidemiologia , Sistema Renina-Angiotensina/fisiologia , Trombofilia/sangue , Trombofilia/epidemiologia , Trombofilia/imunologia , Trombose/sangue , Trombose/imunologia , Tratamento Farmacológico da COVID-19
17.
Clin Appl Thromb Hemost ; 26: 1076029620943293, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32735131

RESUMO

Since the onset of the global pandemic in early 2020, coronavirus disease 2019 (COVID-19) has posed a multitude of challenges to health care systems worldwide. In order to combat these challenges and devise appropriate therapeutic strategies, it becomes of paramount importance to elucidate the pathophysiology of this illness. Coronavirus disease 2019, caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV2), is characterized by a dysregulated immune system and hypercoagulability. COVID-associated coagulopathy (CAC) was recognized based on profound d-dimer elevations and evidence of microthrombi and macrothrombi, both in venous and arterial systems. The underlying mechanisms associated with CAC have been suggested, but not clearly defined. The model of immunothrombosis illustrates the elaborate crosstalk between the innate immune system and coagulation. The rendering of a procoagulant state in COVID-19 involves the interplay of many innate immune pathways. The SARS-CoV2 virus can directly infect immune and endothelial cells, leading to endothelial injury and dysregulation of the immune system. Activated leukocytes potentiate a procoagulant state via release of intravascular tissue factor, platelet activation, NETosis, and inhibition of anticoagulant mechanisms. Additional pathways of specific relevance in CAC include cytokine release and complement activation. All these mechanisms have recently been reported in COVID-19. Immunothrombosis provides a comprehensive perspective of the several synergistic pathways pertinent to the pathogenesis of CAC.


Assuntos
Betacoronavirus , Transtornos da Coagulação Sanguínea/virologia , Infecções por Coronavirus/complicações , Pneumonia Viral/complicações , Transtornos da Coagulação Sanguínea/etiologia , Transtornos da Coagulação Sanguínea/patologia , COVID-19 , Infecções por Coronavirus/fisiopatologia , Infecções por Coronavirus/virologia , Células Endoteliais/patologia , Células Endoteliais/virologia , Humanos , Imunidade Inata , Leucócitos/metabolismo , Leucócitos/patologia , Pandemias , Pneumonia Viral/fisiopatologia , Pneumonia Viral/virologia , SARS-CoV-2 , Trombofilia/imunologia , Trombofilia/virologia , Trombose/etiologia , Trombose/imunologia , Trombose/virologia
18.
Life Sci ; 258: 118166, 2020 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-32739471

RESUMO

In this paper, a model is proposed of the pathophysiological processes of COVID-19 starting from the infection of human type II alveolar epithelial cells (pneumocytes) by SARS-CoV-2 and culminating in the development of ARDS. The innate immune response to infection of type II alveolar epithelial cells leads both to their death by apoptosis and pyroptosis and to alveolar macrophage activation. Activated macrophages secrete proinflammatory cytokines and chemokines and tend to polarise into the inflammatory M1 phenotype. These changes are associated with activation of vascular endothelial cells and thence the recruitment of highly toxic neutrophils and inflammatory activated platelets into the alveolar space. Activated vascular endothelial cells become a source of proinflammatory cytokines and reactive oxygen species (ROS) and contribute to the development of coagulopathy, systemic sepsis, a cytokine storm and ARDS. Pulmonary activated platelets are also an important source of proinflammatory cytokines and ROS, as well as exacerbating pulmonary neutrophil-mediated inflammatory responses and contributing to systemic sepsis by binding to neutrophils to form platelet-neutrophil complexes (PNCs). PNC formation increases neutrophil recruitment, activation priming and extraversion of these immune cells into inflamed pulmonary tissue, thereby contributing to ARDS. Sequestered PNCs cause the development of a procoagulant and proinflammatory environment. The contribution to ARDS of increased extracellular histone levels, circulating mitochondrial DNA, the chromatin protein HMGB1, decreased neutrophil apoptosis, impaired macrophage efferocytosis, the cytokine storm, the toll-like receptor radical cycle, pyroptosis, necroinflammation, lymphopenia and a high Th17 to regulatory T lymphocyte ratio are detailed.


Assuntos
Betacoronavirus/fisiologia , Infecções por Coronavirus/fisiopatologia , Pneumonia Viral/fisiopatologia , Síndrome do Desconforto Respiratório/fisiopatologia , Células Epiteliais Alveolares/imunologia , Células Epiteliais Alveolares/patologia , Animais , Betacoronavirus/imunologia , COVID-19 , Infecções por Coronavirus/complicações , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/terapia , Humanos , Imunidade Inata , Inflamação/etiologia , Inflamação/imunologia , Inflamação/fisiopatologia , Inflamação/terapia , Ativação de Macrófagos , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/patologia , Ativação de Neutrófilo , Pandemias , Ativação Plaquetária , Pneumonia Viral/complicações , Pneumonia Viral/imunologia , Pneumonia Viral/terapia , Síndrome do Desconforto Respiratório/etiologia , Síndrome do Desconforto Respiratório/imunologia , Síndrome do Desconforto Respiratório/terapia , SARS-CoV-2 , Trombofilia/etiologia , Trombofilia/imunologia , Trombofilia/fisiopatologia , Trombofilia/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA