Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
2.
J Autoimmun ; 143: 103166, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38219652

RESUMO

The complement system plays a central role in the pathogenesis of Systemic Lupus Erythematosus (SLE), but most studies have focused on the classical pathway. Ficolin-3 is the main initiator of the lectin pathway of complement in humans, but its role in systemic autoimmune disease has not been conclusively determined. Here, we combined biochemical and genetic approaches to assess the contribution of ficolin-3 to SLE risk and disease manifestations. Ficolin-3 activity was measured by a functional assay in serum or plasma samples from Swedish SLE patients (n = 786) and controls matched for age and sex (n = 566). Genetic variants in an extended 300 kb genomic region spanning the FCN3 locus were analyzed for their association with ficolin-3 activity and SLE manifestations in a Swedish multicenter cohort (n = 985). Patients with ficolin-3 activity in the highest tertile showed a strong enrichment in an SLE cluster defined by anti-Sm/DNA/nucleosome antibodies (OR 3.0, p < 0.001) and had increased rates of hematological disease (OR 1.4, p = 0.078) and lymphopenia (OR = 1.6, p = 0.039). Genetic variants associated with low ficolin-3 activity mapped to an extended haplotype in high linkage disequilibrium upstream of the FCN3 gene. Patients carrying the lead genetic variant associated with low ficolin-3 activity had a lower frequency of hematological disease (OR 0.67, p = 0.018) and lymphopenia (OR 0.63, p = 0.031) and fewer autoantibodies (p = 0.0019). Loss-of-function variants in the FCN3 gene were not associated with SLE, but four (0.5 %) SLE patients developed acquired ficolin-3 deficiency where ficolin-3 activity in serum was depleted following diagnosis of SLE. Taken together, our results provide genetic and biochemical evidence that implicate the lectin pathway in hematological SLE manifestations. We also identify lectin pathway activation through ficolin-3 as a factor that contributes to the autoantibody response in SLE.


Assuntos
Doenças Hematológicas , Lúpus Eritematoso Sistêmico , Linfopenia , Humanos , Anticorpos Antinucleares , Autoanticorpos , Proteínas do Sistema Complemento , Ficolinas , Lectinas/genética , Lúpus Eritematoso Sistêmico/diagnóstico , Lúpus Eritematoso Sistêmico/epidemiologia , Lúpus Eritematoso Sistêmico/genética
3.
J Biol Chem ; 294(13): 4878-4888, 2019 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-30670593

RESUMO

ER protein 57 (ERp57), a thiol isomerase secreted from vascular cells, is essential for complete thrombus formation in vivo, but other extracellular ERp57 functions remain unexplored. Here, we employed a kinetic substrate-trapping approach to identify extracellular protein substrates of ERp57 in platelet-rich plasma. MS-based identification with immunochemical confirmation combined with gene ontology enrichment analysis revealed that ERp57 targets, among other substrates, components of the lectin pathway of complement activation: mannose-binding lectin, ficolin-2, ficolin-3, collectin-10, collectin-11, mannose-binding lectin-associated serine protease-1, and mannose-binding lectin-associated serine protease-2. Ficolin-3, the most abundant lectin pathway initiator in humans, circulates as disulfide-linked multimers of a monomer. ERp57 attenuated ficolin-3 ligand recognition and complement activation by cleaving intermolecular disulfide bonds in large ficolin-3 multimers, thereby reducing multimer size and ligand-binding affinity. We used MS to identify the disulfide-bonding pattern in ficolin-3 multimers and the disulfide bonds targeted by ERp57 and found that Cys6 and Cys23 in the N-terminal region of ficolin-3 form the intermolecular disulfide bonds in ficolin-3 multimers that are reduced by ERp57. Our results not only demonstrate that ERp57 can negatively regulate complement activation, but also identify a control mechanism for lectin pathway initiation in the vasculature. We conclude that extensive multimerization in large ficolin-3 multimers leads to a high affinity for ligands and strong complement-activating potential and that ERp57 suppresses complement activation by cleaving disulfide bonds in ficolin-3 and reducing its multimer size.


Assuntos
Lectina de Ligação a Manose da Via do Complemento , Glicoproteínas/metabolismo , Lectinas/metabolismo , Isomerases de Dissulfetos de Proteínas/metabolismo , Multimerização Proteica , Proteólise , Glicoproteínas/genética , Humanos , Lectinas/genética , Isomerases de Dissulfetos de Proteínas/genética
4.
J Med Internet Res ; 22(11): e21559, 2020 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-33031049

RESUMO

BACKGROUND: The COVID-19 pandemic has spread at an alarming speed, and effective treatment for the disease is still lacking. The body of evidence on COVID-19 has been increasing at an impressive pace, creating the need for a method to rapidly assess the current knowledge and identify key information. Gold standard methods such as systematic reviews and meta-analyses are regarded unsuitable because they have a narrow scope and are very time consuming. OBJECTIVE: This study aimed to explore the published scientific literature on COVID-19 and map the research evolution during the early phase of the COVID-19 pandemic. METHODS: We performed a PubMed search to analyze the titles, keywords, and abstracts of published papers on COVID-19. We used latent Dirichlet allocation modeling to extract topics and conducted a trend analysis to understand the temporal changes in research for each topic, journal impact factor (JIF), and geographic origin. RESULTS: Based on our search, we identified 16,670 relevant articles dated between February 14, 2020, and June 1, 2020. Of these, 6 articles were reports from peer-reviewed randomized trials on patients with COVID-19. We identified 14 main research topics, of which the most common topics were health care responses (2812/16,670, 16.86%) and clinical manifestations (1828/16,670, 10.91%). We found an increasing trend for research on clinical manifestations and protective measures and a decreasing trend for research on disease transmission, epidemiology, health care response, and radiology. Publications on protective measures, immunology, and clinical manifestations were associated with the highest JIF. The overall median JIF was 3.7 (IQR 2.6-5.9), and we found that the JIF for these publications declined over time. The top countries producing research were the United States, China, Italy, and the United Kingdom. CONCLUSIONS: In less than 6 months since the novel coronavirus was first detected, a remarkably high number of research articles on COVID-19 have been published. Here, we discuss and present the temporal changes in the available COVID-19 research during the early phase of the pandemic. Our findings may aid researchers and policy makers to form a structured view of the current COVID-19 evidence base and provide further research directions.


Assuntos
COVID-19/epidemiologia , Publicações/normas , SARS-CoV-2/patogenicidade , Humanos , Projetos de Pesquisa
6.
BMC Cancer ; 16: 341, 2016 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-27246245

RESUMO

BACKGROUND: Tissue Factor (TF) forms a proteolytically active complex together with coagulation factor VIIa (FVIIa) and functions as the trigger of blood coagulation or alternatively activates cell signaling. We recently described that EphA2 of the Eph tyrosine kinase receptor family is cleaved directly by the TF/FVIIa complex. The aim of the present study was to further characterize the cross-talk between TF/FVIIa and EphA2 using in vitro model systems and human cancer specimens. METHODS: Cleavage and phosphorylation of EphA2 was studied by Western blot. Subcellular localization of TF and EphA2 was investigated by a proximity ligation assay and confocal microscopy. Phalloidin staining of the actin cytoskeleton was used to study cell rounding and retraction fiber formation. Expression of TF and EphA2 in human colorectal cancer specimens was examined by immunohistochemistry. RESULTS: TF and EphA2 co-localized constitutively in MDA-MB-231 cells, and addition of FVIIa resulted in cleavage of EphA2 by a PAR2-independent mechanism. Overexpression of TF in U251 glioblastoma cells lead to co-localization with EphA2 at the leading edge and FVIIa-dependent cleavage of EphA2. FVIIa potentiated ephrin-A1-induced cell rounding and retraction fiber formation in MDA-MB-231 cells through a RhoA/ROCK-dependent pathway that did not require PAR2-activation. TF and EphA2 were expressed in colorectal cancer specimens, and were significantly correlated. CONCLUSIONS: These results suggest that TF/FVIIa-EphA2 cross-talk might potentiate ligand-dependent EphA2 signaling in human cancers, and provide initial evidence that it is possible for this interaction to occur in vivo.


Assuntos
Neoplasias Colorretais/metabolismo , Fator VIIa/metabolismo , Receptor Cross-Talk/fisiologia , Receptor EphA2/metabolismo , Tromboplastina/metabolismo , Western Blotting , Técnicas de Silenciamento de Genes , Humanos , Imuno-Histoquímica , Microscopia Confocal , Análise Serial de Tecidos
8.
J Biol Chem ; 289(47): 32379-91, 2014 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-25281742

RESUMO

Tissue factor (TF) binds the serine protease factor VIIa (FVIIa) to form a proteolytically active complex that can trigger coagulation or activate cell signaling. Here we addressed the involvement of tyrosine kinase receptors (RTKs) in TF/FVIIa signaling by antibody array analysis and subsequently found that EphB2 and EphA2 of the Eph RTK family were cleaved in their ectodomains by TF/FVIIa. We used N-terminal Edman sequencing and LC-MS/MS analysis to characterize the cleaved Eph isoforms and identified a key arginine residue at the cleavage site, in agreement with the tryptic serine protease activity of FVIIa. Protease-activated receptor 2 (PAR2) signaling and downstream coagulation activity was non-essential in this context, in further support of a direct cleavage by TF/FVIIa. EphB2 was cleaved by FVIIa concentrations in the subnanomolar range in a number of TF expressing cell types, indicating that the active cellular pool of TF was involved. FVIIa caused potentiation of cell repulsion by the EphB2 ligand ephrin-B1, demonstrating a novel proteolytical event to control Eph-mediated cell segregation. These results define Eph RTKs as novel proteolytical targets of TF/FVIIa and provide new insights into how TF/FVIIa regulates cellular functions independently of PAR2.


Assuntos
Fator VIIa/metabolismo , Receptor EphA2/metabolismo , Receptor EphB2/metabolismo , Tromboplastina/metabolismo , Sequência de Aminoácidos , Sítios de Ligação/genética , Western Blotting , Linhagem Celular Tumoral , Movimento Celular , Células Cultivadas , Dissulfetos/química , Dissulfetos/metabolismo , Fator VII , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Oxirredução , Ligação Proteica , Estrutura Terciária de Proteína , Proteólise , Receptor EphA2/química , Receptor EphA2/genética , Receptor EphB2/química , Receptor EphB2/genética , Análise de Sequência de Proteína , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Especificidade por Substrato , Espectrometria de Massas em Tandem
9.
Semin Thromb Hemost ; 41(7): 691-9, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26418256

RESUMO

Tissue factor (TF) is a 47-kDa transmembrane glycoprotein and the main initiator of the blood coagulation cascade. Binding to its ligand factor VIIa (FVIIa) also initiates noncoagulant signaling with broad biological implications. In this review, we discuss how TF interacts with other cell-surface proteins, which affect biological functions such as cell migration and cell survival. A vast number of publications have demonstrated the importance of TF-induced activation of protease-activated receptors, but recently published research has indicated a more complicated picture. As it has been discovered that TF interacts with integrins and receptor tyrosine kinases, novel signaling mechanisms for the TF/FVIIa complex have been presented. The knowledge of these new aspects of TF signaling may, for instance, facilitate the development of new treatment strategies for cancer and acute coronary syndromes, two examples of diseases characterized by aberrant TF expression and signaling.


Assuntos
Síndrome Coronariana Aguda/metabolismo , Apoptose , Movimento Celular , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Transdução de Sinais , Tromboplastina/metabolismo , Síndrome Coronariana Aguda/patologia , Síndrome Coronariana Aguda/terapia , Animais , Sobrevivência Celular , Fator VIIa/metabolismo , Humanos , Neoplasias/patologia , Neoplasias/terapia , Ligação Proteica
11.
Scand J Clin Lab Invest ; 75(5): 428-33, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25874478

RESUMO

OBJECTIVE: To evaluate the Trillium Diagnostics Leuko64(™) assay on Abbott Celldyn Sapphire haematology analyser compared to two flow cytometry protocols on Beckman Coulter EPICS MCL flow cytometer. MATERIALS AND METHODS: CD64 expression on neutrophils was determined by two flow cytometry protocols and by a commercial assay on an automatic haematology analyser. The inclusion of study subjects was based on elevated procalcitonin (PCT) values, identifying patients where a systemic infection was suspected. Healthy blood donors were used as a reference group. RESULTS: Statistically significant correlations between the Trillium Diagnostics Leuko64(™) assay and the flow cytometry methods were found when measuring neutrophil CD64 expression. CONCLUSIONS: The good correlation between a reference method and an automated haematology analyser method for CD64 expression on neutrophils supports introduction of the latter assay for routine use as an independent biomarker of bacterial infection and inflammation.


Assuntos
Citometria de Fluxo/instrumentação , Citometria de Fluxo/métodos , Testes Hematológicos/instrumentação , Neutrófilos/metabolismo , Kit de Reagentes para Diagnóstico , Receptores de IgG/metabolismo , Intervalos de Confiança , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Padrões de Referência
12.
Front Psychol ; 14: 1196945, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37744585

RESUMO

Introduction: Repetitive negative thinking (RNT) has been described as a maintaining transdiagnostic factor for psychopathology within the areas of depression, anxiety and insomnia. We investigated the effects of rumination-focused cognitive-behaviour therapy (RF-CBT) in a group format at a primary health care centre on symptoms of depression, anxiety, insomnia, RNT, and quality of life. The participants presented clinical symptom levels of worry and at least two disorders among anxiety disorders, major depressive disorder, and insomnia disorder. Methods: A randomised controlled superiority parallel arm trial was used. 73 participants were included and randomised in pairs to either group-administered RF-CBT or a waiting list condition. The primary outcomes were self-rated worry and transdiagnostic symptoms (depression, anxiety, and insomnia). Intention-to-treat analyses of group differences were conducted using linear mixed models. Adverse side effects and incidents were presented descriptively. Results: Group RF-CBT significantly reduced self-reported insomnia at post-treatment and self-reported insomnia and depression at the 2 month-follow-up, relative to the wait-list control group. There was no significant difference in change in RNT, anxiety, or quality of life. Discussion: The current study suggests that group-administered RF-CBT may be effective for insomnia and potentially effective for depression symptomatology. However, the study was underpowered to detect small and moderate effects and the results should therefore be interpreted with caution.

13.
J Innate Immun ; 15(1): 412-427, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36858027

RESUMO

The complement system plays a crucial role in host defense, homeostasis, and tissue regeneration and bridges the innate and the adaptive immune systems. Although the genetic variants in complement C2 (c.839_849+17del; p.(Met280Asnfs*5)) and C8B (c.1625C>T; p.(Thr542Ile)) are known individually, here, we report on a patient carrying their combination in a heterozygous form. The patient presented with a reduced general condition and suffers from a wide variety of autoimmune diseases. While no autoimmune disease-specific autoantibodies could be detected, genetic analysis revealed abnormalities in the two complement genes C2 and C8B. Therefore, we performed a comprehensive investigation of the innate immune system on a cellular and humoral level to define the functional consequences. We found slightly impaired functionality of neutrophils and monocytes regarding phagocytosis and reactive oxygen species generation and a diminished expression of the C5aR1. An extensive complement analysis revealed a declined activation potential for the alternative and classical pathway. Reconstitution with purified C2 and C8 into patient serum failed to normalize the dysfunction, whereas the addition of C3 improved the hemolytic activity. In clinical transfer, in vitro supplementation of the patient's plasma with FFP as a complement source could fully restore full complement functionality. This study describes for the first time a combined heterozygous genetic variation in complement C2 and C8B which, however, cannot fully explain the overall dysfunctions and calls for further complement deficiency research and corresponding therapies.


Assuntos
Doenças Autoimunes , Complemento C2 , Humanos , Ativação do Complemento/genética , Complemento C2/genética , Proteínas do Sistema Complemento/genética , Variação Genética/genética
14.
Blood Adv ; 7(20): 6367-6380, 2023 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-37428869

RESUMO

Complement activation in the diseases paroxysmal nocturnal hemoglobinuria (PNH) and atypical hemolytic uremic syndrome (aHUS) results in cytolysis and fatal thrombotic events, which are largely refractory to anticoagulation and/or antiplatelet therapy. Anticomplement therapy, however, efficiently prevents thrombotic events in PNH and aHUS, but the underlying mechanisms remain unresolved. We show that complement-mediated hemolysis in whole blood induces platelet activation similarly to activation by adenosine 5'-diphosphate (ADP). Blockage of C3 or C5 abolished platelet activation. We found that human platelets failed to respond functionally to the anaphylatoxins C3a and C5a. Instead, complement activation did lead to prothrombotic cell activation in the whole blood when membrane attack complex (MAC)-mediated cytolysis occurred. Consequently, we demonstrate that ADP receptor antagonists efficiently inhibited platelet activation, although full complement activation, which causes hemolysis, occurred. By using an established model of mismatched erythrocyte transfusions in rats, we crossvalidated these findings in vivo using the complement inhibitor OmCI and cobra venom factor. Consumptive complement activation in this animal model only led to a thrombotic phenotype when MAC-mediated cytolysis occurred. In conclusion, complement activation only induces substantial prothrombotic cell activation if terminal pathway activation culminates in MAC-mediated release of intracellular ADP. These results explain why anticomplement therapy efficiently prevents thromboembolisms without interfering negatively with hemostasis.


Assuntos
Síndrome Hemolítico-Urêmica Atípica , Hemoglobinúria Paroxística , Humanos , Ratos , Animais , Complexo de Ataque à Membrana do Sistema Complemento , Hemólise , Eritrócitos/metabolismo , Ativação do Complemento , Plaquetas/metabolismo , Hemoglobinúria Paroxística/genética
15.
Med Phys ; 49(6): 3564-3573, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35305023

RESUMO

PURPOSE: We present a framework for robust automated treatment planning using machine learning, comprising scenario-specific dose prediction and robust dose mimicking. METHODS: The scenario dose prediction pipeline is divided into the prediction of nominal dose from input image and the prediction of scenario dose from nominal dose, each using a deep learning model with U-net architecture. By using a specially developed dose-volume histogram-based loss function, the predicted scenario doses are ensured sufficient target coverage despite the possibility of the training data being non-robust. Deliverable plans may then be created by solving a robust dose mimicking problem with the predictions as scenario-specific reference doses. RESULTS: Numerical experiments are performed using a data set of 52 intensity-modulated proton therapy plans for prostate patients. We show that the predicted scenario doses resemble their respective ground truth well, in particular while having target coverage comparable to that of the nominal scenario. The deliverable plans produced by the subsequent robust dose mimicking were showed to be robust against the same scenario set considered for prediction. CONCLUSIONS: We demonstrate the feasibility and merits of the proposed methodology for incorporating robustness into automated treatment planning algorithms.


Assuntos
Terapia com Prótons , Radioterapia de Intensidade Modulada , Humanos , Masculino , Órgãos em Risco , Terapia com Prótons/métodos , Dosagem Radioterapêutica , Planejamento da Radioterapia Assistida por Computador/métodos , Radioterapia de Intensidade Modulada/métodos
16.
Front Immunol ; 13: 1030627, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36820001

RESUMO

COVID-19 has been shown to have a multifaceted impact on the immune system. In a recently published article in Front Immunol, we show that the intravascular innate immune system (IIIS) is strongly activated in severe COVID-19 with ARDS and appears to be one of the causes leading to severe COVID-19. In this article, we describe the IIIS and its physiological function, but also the strong pro-inflammatory effects that are observed in COVID-19 and in various other pathological conditions and treatments such as during ischemia reperfusion injury and in treatments where biomaterials come in direct contact with blood in, e.g., extracorporeal and intravasal treatments. In the present article, we describe how the IIIS, a complex network of plasma proteins and blood cells, constitute the acute innate immune response of the blood and discuss the effects that the IIIS induces in pathological disorders and treatments in modern medicine.


Assuntos
COVID-19 , Humanos , Imunidade Inata , Sistema Imunitário
17.
Front Immunol ; 13: 840137, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35350780

RESUMO

Most SARS-CoV-2 infected patients experience influenza-like symptoms of low or moderate severity. But, already in 2020 early during the pandemic it became obvious that many patients had a high incidence of thrombotic complications, which prompted treatment with high doses of low-molecular-weight heparin (LMWH; typically 150-300IU/kg) to prevent thrombosis. In some patients, the disease aggravated after approximately 10 days and turned into a full-blown acute respiratory distress syndrome (ARDS)-like pulmonary inflammation with endothelialitis, thrombosis and vascular angiogenesis, which often lead to intensive care treatment with ventilator support. This stage of the disease is characterized by dysregulation of cytokines and chemokines, in particular with high IL-6 levels, and also by reduced oxygen saturation, high risk of thrombosis, and signs of severe pulmonary damage with ground glass opacities. The direct link between SARS-CoV-2 and the COVID-19-associated lung injury is not clear. Indirect evidence speaks in favor of a thromboinflammatory reaction, which may be initiated by the virus itself and by infected damaged and/or apoptotic cells. We and others have demonstrated that life-threatening COVID-19 ARDS is associated with a strong activation of the intravascular innate immune system (IIIS). In support of this notion is that activation of the complement and kallikrein/kinin (KK) systems predict survival, the necessity for usage of mechanical ventilation, acute kidney injury and, in the case of MBL, also coagulation system activation with thromboembolism. The general properties of the IIIS can easily be translated into mechanisms of COVID-19 pathophysiology. The prognostic value of complement and KKsystem biomarkers demonstrate that pharmaceuticals, which are licensed or have passed the phase I trial stage are promising candidate drugs for treatment of COVID-19. Examples of such compounds include complement inhibitors AMY-101 and eculizumab (targeting C3 and C5, respectively) as well as kallikrein inhibitors ecallantide and lanadelumab and the bradykinin receptor (BKR) 2 antagonist icatibant. In this conceptual review we discuss the activation, crosstalk and the therapeutic options that are available for regulation of the IIIS.


Assuntos
COVID-19 , Síndrome do Desconforto Respiratório , Trombose , COVID-19/complicações , Heparina de Baixo Peso Molecular/uso terapêutico , Humanos , Sistema Imunitário , Calicreínas , Síndrome do Desconforto Respiratório/etiologia , Síndrome do Desconforto Respiratório/terapia , SARS-CoV-2 , Trombose/tratamento farmacológico
18.
J Clin Med ; 11(12)2022 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-35743491

RESUMO

PURPOSE: the pathophysiologic mechanisms explaining differences in clinical outcomes following COVID-19 are not completely described. This study aims to investigate antibody responses in critically ill patients with COVID-19 in relation to inflammation, organ failure and 30-day survival. METHODS: All patients with PCR-verified COVID-19 and gave consent, and who were admitted to a tertiary Intensive care unit (ICU) in Sweden during March-September 2020 were included. Demography, repeated blood samples and measures of organ function were collected. Analyses of anti-SARS-CoV-2 antibodies (IgM, IgA and IgG) in plasma were performed and correlated to patient outcome and biomarkers of inflammation and organ failure. RESULTS: A total of 115 patients (median age 62 years, 77% male) were included prospectively. All patients developed severe respiratory dysfunction, and 59% were treated with invasive ventilation. Thirty-day mortality was 22.6% for all included patients. Patients negative for any anti-SARS-CoV-2 antibody in plasma during ICU admission had higher 30-day mortality compared to patients positive for antibodies. Patients positive for IgM had more ICU-, ventilator-, renal replacement therapy- and vasoactive medication-free days. IgA antibody concentrations correlated negatively with both SAPS3 and maximal SOFA-score and IgM-levels correlated negatively with SAPS3. Patients with antibody levels below the detection limit had higher plasma levels of extracellular histones on day 1 and elevated levels of kidney and cardiac biomarkers, but showed no signs of increased inflammation, complement activation or cytokine release. After adjusting for age, positive IgM and IgG antibodies were still associated with increased 30-day survival, with odds ratio (OR) 7.1 (1.5-34.4) and 4.2 (1.1-15.7), respectively. CONCLUSION: In patients with severe COVID-19 requiring intensive care, a poor antibody response is associated with organ failure, systemic histone release and increased 30-day mortality.

19.
Front Immunol ; 12: 627579, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33692801

RESUMO

An important manifestation of severe COVID-19 is the ARDS-like lung injury that is associated with vascular endothelialitis, thrombosis, and angiogenesis. The intravascular innate immune system (IIIS), including the complement, contact, coagulation, and fibrinolysis systems, which is crucial for recognizing and eliminating microorganisms and debris in the body, is likely to be involved in the pathogenesis of COVID-19 ARDS. Biomarkers for IIIS activation were studied in the first 66 patients with COVID-19 admitted to the ICU in Uppsala University Hospital, both cross-sectionally on day 1 and in 19 patients longitudinally for up to a month, in a prospective study. IIIS analyses were compared with biochemical parameters and clinical outcome and survival. Blood cascade systems activation leading to an overreactive conjunct thromboinflammation was demonstrated, reflected in consumption of individual cascade system components, e.g., FXII, prekallikrein, and high molecular weight kininogen and in increased levels of activation products, e.g., C4d, C3a, C3d,g, sC5b-9, TAT, and D-dimer. Strong associations were found between the blood cascade systems and organ damage, illness severity scores, and survival. We show that critically ill COVID-19 patients display a conjunct activation of the IIIS that is linked to organ damage of the lung, heart, kidneys, and death. We present evidence that the complement and in particular the kallikrein/kinin system is strongly activated and that both systems are prognostic markers of the outcome of the patients suggesting their role in driving the inflammation. Already licensed kallikrein/kinin inhibitors are potential drugs for treatment of critically ill patients with COVID-19.


Assuntos
COVID-19/imunologia , COVID-19/metabolismo , Inflamação/imunologia , Sistema Calicreína-Cinina/imunologia , Trombose/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/metabolismo , Coagulação Sanguínea , COVID-19/patologia , COVID-19/virologia , Estado Terminal , Feminino , Fibrinólise/imunologia , Humanos , Imunidade Inata , Inflamação/metabolismo , Inflamação/patologia , Inflamação/virologia , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Síndrome do Desconforto Respiratório/imunologia , Síndrome do Desconforto Respiratório/metabolismo , Síndrome do Desconforto Respiratório/patologia , Síndrome do Desconforto Respiratório/virologia , SARS-CoV-2/isolamento & purificação , Índice de Gravidade de Doença , Adulto Jovem
20.
Thromb Haemost ; 120(12): 1720-1724, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32871607

RESUMO

The ongoing COVID-19 pandemic has caused significant morbidity and mortality worldwide, as well as profound effects on society. COVID-19 patients have an increased risk of thromboembolic (TE) complications, which develop despite pharmacological thromboprophylaxis. The mechanism behind COVID-19-associated coagulopathy remains unclear. Mannose-binding lectin (MBL), a pattern recognition molecule that initiates the lectin pathway of complement activation, has been suggested as a potential amplifier of blood coagulation during thromboinflammation. Here we describe data from a cohort of critically ill COVID-19 patients (n = 65) treated at a tertiary hospital center intensive care unit (ICU). A subset of patients had strongly elevated MBL plasma levels, and activity upon ICU admission, and patients who developed symptomatic TE (14%) had significantly higher MBL levels than patients without TE. MBL was strongly correlated to plasma D-dimer levels, a marker of COVID-19 coagulopathy, but showed no relationship to degree of inflammation or other organ dysfunction. In conclusion, we have identified complement activation through the MBL pathway as a novel amplification mechanism that contributes to pathological thrombosis in critically ill COVID-19 patients. Pharmacological targeting of the MBL pathway could be a novel treatment option for thrombosis in COVID-19. Laboratory testing of MBL levels could be of value for identifying COVID-19 patients at risk for TE events.


Assuntos
Biomarcadores/sangue , Transtornos da Coagulação Sanguínea/diagnóstico por imagem , COVID-19/diagnóstico , Estado Terminal , Lectina de Ligação a Manose/sangue , SARS-CoV-2/fisiologia , Tromboembolia Venosa/diagnóstico , Adulto , Idoso , Idoso de 80 Anos ou mais , Ativação do Complemento , Feminino , Produtos de Degradação da Fibrina e do Fibrinogênio/metabolismo , Humanos , Unidades de Terapia Intensiva , Masculino , Pessoa de Meia-Idade , Pandemias , Risco , Suécia , Centros de Atenção Terciária , Regulação para Cima , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA