Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
EBioMedicine ; 75: 103812, 2022 Jan.
Article in English | MEDLINE | ID: covidwho-1639102

ABSTRACT

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.


Subject(s)
Blood Coagulation Disorders/metabolism , COVID-19/metabolism , Down-Regulation , Endothelium, Vascular/metabolism , Hypoxia/metabolism , Lung/metabolism , SARS-CoV-2/metabolism , Thrombomodulin/biosynthesis , Aged , Aged, 80 and over , Blood Coagulation Disorders/pathology , COVID-19/pathology , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Endothelium, Vascular/ultrastructure , Female , Humans , Hypoxia/pathology , Lung/ultrastructure , Male , Middle Aged
2.
Cell Mol Biol Lett ; 27(1): 6, 2022 Jan 11.
Article in English | MEDLINE | ID: covidwho-1622208

ABSTRACT

Coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), is associated with a high mortality rate. The majority of deaths in this disease are caused by ARDS (acute respiratory distress syndrome) followed by cytokine storm and coagulation complications. Although alterations in the level of the number of coagulation factors have been detected in samples from COVID-19 patients, the direct molecular mechanism which has been involved in this pathologic process has not been explored yet. The PI3K/AKT signaling pathway is an intracellular pathway which plays a central role in cell survival. Also, in recent years the association between this pathway and coagulopathies has been well clarified. Therefore, based on the evidence on over-activity of the PI3K/AKT signaling pathway in SARS-CoV-2 infection, in the current review, the probable role of this cellular pathway as a therapeutic target for the prevention of coagulation complications in patients with COVID-19 is discussed.


Subject(s)
Blood Coagulation Disorders/etiology , Blood Coagulation , COVID-19/complications , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Animals , Blood Coagulation Disorders/blood , Blood Coagulation Disorders/metabolism , COVID-19/blood , COVID-19/metabolism , Humans , Inflammation/blood , Inflammation/etiology , Inflammation/metabolism , Molecular Targeted Therapy , SARS-CoV-2/physiology
3.
Am J Physiol Heart Circ Physiol ; 322(1): H87-H93, 2022 01 01.
Article in English | MEDLINE | ID: covidwho-1566407

ABSTRACT

The 2019 coronavirus disease (COVID-19) is the disease caused by SARS-CoV-2 infection. Although this infection has been shown to affect the respiratory system, a high incidence of thrombotic events has been observed in severe cases of COVID-19 and in a significant portion of COVID-19 nonsurvivors. Although prior literature has reported on both the coagulopathy and hypercoagulability of COVID-19, the specifics of coagulation have not been fully investigated. Observations of microthrombosis in patients with COVID-19 have brought attention to potential inflammatory endothelial injury. Von Willebrand factor (VWF) and its protease, A disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13 (ADAMTS13), play an important homeostatic role in responding to endothelial injury. This report provides an overview of the literature investigating the role the VWF/ADAMTS13 axis may have in COVID-19 thrombotic events and suggests potential therapeutic strategies to prevent the progression of coagulopathy in patients with COVID-19.


Subject(s)
ADAMTS13 Protein/metabolism , Blood Coagulation Disorders/metabolism , COVID-19/blood , von Willebrand Factor/metabolism , Blood Coagulation , Blood Coagulation Disorders/etiology , COVID-19/complications , Humans
4.
Clin Immunol ; 232: 108852, 2021 11.
Article in English | MEDLINE | ID: covidwho-1401324

ABSTRACT

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.


Subject(s)
Blood Coagulation/physiology , COVID-19/complications , Fibrinolysis/physiology , Inflammation/etiology , Inflammation/virology , Adult , Anticoagulants/pharmacology , Blood Coagulation/drug effects , Blood Coagulation Disorders/etiology , Blood Coagulation Disorders/metabolism , Blood Coagulation Disorders/pathology , Blood Coagulation Disorders/virology , COVID-19/metabolism , COVID-19/pathology , China , Disease Progression , Disseminated Intravascular Coagulation/etiology , Disseminated Intravascular Coagulation/metabolism , Disseminated Intravascular Coagulation/pathology , Disseminated Intravascular Coagulation/virology , Female , Fibrin Fibrinogen Degradation Products/metabolism , Fibrinogen/metabolism , Hemorrhage/etiology , Hemorrhage/pathology , Hemorrhage/virology , Humans , Inflammation/pathology , Male , Middle Aged , Prothrombin Time , SARS-CoV-2/pathogenicity
5.
Front Immunol ; 12: 649122, 2021.
Article in English | MEDLINE | ID: covidwho-1285287

ABSTRACT

Thromboplasminflammation in coronavirus disease 2019 (COVID-19) coagulopathy consists of angiotensin II (Ang II)-induced coagulopathy, activated factor XII (FXIIa)- and kallikrein, kinin system-enhanced fibrinolysis, and disseminated intravascular coagulation (DIC). All three conditions induce systemic inflammation via each pathomechanism-developed production of inflammatory cytokines. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) downregulates angiotensin-converting enzyme 2, leading to an increase in Ang II levels. Ang II-induced coagulopathy comprising platelet activation, thrombin generation, plasminogen activator inhibitor-1 expression and endothelial injury causes thrombosis via the angiotensin II type 1 receptor. SARS-CoV-2 RNA and neutrophil extracellular trap (NET) DNA activate FXII, resulting in plasmin generation through FXIIa- and kallikrein-mediated plasminogen conversion to plasmin and bradykinin-induced tissue-type plasminogen activator release from the endothelium via the kinin B2 receptor. NETs induce immunothrombosis at the site of infection (lungs), through histone- and DNA-mediated thrombin generation, insufficient anticoagulation control, and inhibition of fibrinolysis. However, if the infection is sufficiently severe, immunothrombosis disseminates into the systemic circulation, and DIC, which is associated with the endothelial injury, occurs. Inflammation, and serine protease networks of coagulation and fibrinolysis, militate each other through complement pathways, which exacerbates three pathologies of COVID-19 coagulopathy. COVID-19 coagulopathy causes microvascular thrombosis and bleeding, resulting in multiple organ dysfunction and death in critically ill patients. Treatment targets for improving the prognosis of COVID-19 coagulopathy include thrombin, plasmin, and inflammation, and SARS-CoV-2 infection. Several drugs are candidates for controlling these conditions; however, further advances are required to establish robust treatments based on a clear understanding of molecular mechanisms of COVID-19 coagulopathy.


Subject(s)
Blood Coagulation Disorders/metabolism , COVID-19/metabolism , SARS-CoV-2/physiology , Angiotensin II/metabolism , Animals , Blood Coagulation Disorders/immunology , COVID-19/immunology , Cytokines/metabolism , Factor XIIa/metabolism , Humans , Inflammation , Inflammation Mediators/metabolism
6.
Am J Respir Cell Mol Biol ; 64(6): 687-697, 2021 06.
Article in English | MEDLINE | ID: covidwho-1143104

ABSTRACT

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.


Subject(s)
Alveolar Epithelial Cells/metabolism , Blood Coagulation Disorders/metabolism , COVID-19/metabolism , Gene Expression Regulation , SARS-CoV-2/metabolism , Signal Transduction , Transcription, Genetic , Alveolar Epithelial Cells/pathology , Alveolar Epithelial Cells/virology , Blood Coagulation Disorders/etiology , Blood Coagulation Disorders/pathology , COVID-19/complications , COVID-19/pathology , Cell Line , Female , Humans , Influenza A virus/metabolism , Influenza, Human/complications , Influenza, Human/metabolism , Influenza, Human/pathology , Male
7.
Adv Respir Med ; 89(1): 90-91, 2021.
Article in English | MEDLINE | ID: covidwho-1073688

ABSTRACT

Since December 2019, the rapid spread of SARS CoV-2 across the border, shuffled into a world pandemic situation with an alarming rate of morbidity and mortality. Concerns are mounting as the reports indicate tangled circumstances among the COVID-19 patients due to blood coagulopathy followed by organ dysfunction. COVID-19 induced an alteration in coagulation function increase the risk of pulmonary embolism and deep vein thrombosis associated with poor prognosis as well as high mortality. An elevated level D-dimer and other fibrin degrading protein are documented among the patients with COVID-19; especially in severe cases. Differences in coagulopathy among severe and non-severe cases, required prompt attention to adopt a more effective management strategy.


Subject(s)
Blood Coagulation Disorders/metabolism , COVID-19/complications , Fibrin Fibrinogen Degradation Products/metabolism , Blood Coagulation Disorders/epidemiology , COVID-19/epidemiology , COVID-19/metabolism , Humans , Incidence , Prognosis , Thrombosis/metabolism
8.
Cardiol Rev ; 29(1): 43-47, 2021.
Article in English | MEDLINE | ID: covidwho-965899

ABSTRACT

The novel coronavirus (severe acute respiratory syndrome CoV-2 [SARS-CoV-2]), also known as COVID-19, is a single-stranded enveloped RNA virus that created a Public Health Emergency of International Concern in January 2020, with a global case burden of over 15 million in just 7 months. Infected patients develop a wide range of clinical manifestations-typically presenting with fever, cough, myalgia, and fatigue. Severely ill patients may fall victim to acute respiratory distress syndrome, acute heart injuries, neurological manifestations, or complications due to secondary infections. These critically ill patients are also found to have disrupted coagulation function, predisposing them to consumptive coagulopathies, and both venous and thromboembolic complications. Common laboratory findings include thrombocytopenia, elevated D-dimer, fibrin degradation products, and fibrinogen, all of which have been associated with greater disease severity. Many cases of pulmonary embolism have been noted, along with deep vein thrombosis, ischemic stroke, myocardial infarction, and systemic arterial embolism. The pathogenesis of coronavirus has not been completely elucidated, but the virus is known to cause excessive inflammation, endothelial injury, hypoxia, and disseminated intravascular coagulation, all of which contribute to thrombosis formation. These patients are also faced with prolonged immobilization while staying in the hospital or intensive care unit. It is important to have a high degree of suspicion for thrombotic complications as patients may rapidly deteriorate in severe cases. Evidence suggests that prophylaxis with anticoagulation may lead to a lower risk of mortality, although it does not eliminate the possibility. The risks and benefits of anticoagulation treatment should be considered in each case. Patients should be regularly evaluated for bleeding risks and thrombotic complications.


Subject(s)
Blood Coagulation Disorders/blood , COVID-19/blood , Embolism/blood , Thrombosis/blood , Anticoagulants/therapeutic use , Blood Coagulation Disorders/drug therapy , Blood Coagulation Disorders/etiology , Blood Coagulation Disorders/metabolism , COVID-19/complications , COVID-19/metabolism , Cytokine Release Syndrome/blood , Cytokine Release Syndrome/complications , Cytokine Release Syndrome/metabolism , Disseminated Intravascular Coagulation/blood , Disseminated Intravascular Coagulation/etiology , Disseminated Intravascular Coagulation/metabolism , Disseminated Intravascular Coagulation/prevention & control , Embolism/etiology , Embolism/metabolism , Embolism/prevention & control , Endothelium, Vascular/metabolism , Fibrin Fibrinogen Degradation Products/metabolism , Fibrinogen/metabolism , Humans , Hypoxia/blood , Hypoxia/etiology , Hypoxia/metabolism , Immobilization , Inflammation/blood , Inflammation/etiology , Inflammation/metabolism , Ischemic Stroke/blood , Ischemic Stroke/etiology , Ischemic Stroke/metabolism , Ischemic Stroke/prevention & control , Myocardial Infarction/blood , Myocardial Infarction/etiology , Myocardial Infarction/metabolism , Myocardial Infarction/prevention & control , Practice Guidelines as Topic , Pulmonary Embolism/blood , Pulmonary Embolism/etiology , Pulmonary Embolism/metabolism , Pulmonary Embolism/prevention & control , Severity of Illness Index , Thrombocytopenia/blood , Thrombocytopenia/etiology , Thrombosis/etiology , Thrombosis/metabolism , Thrombosis/prevention & control , Venous Thrombosis/blood , Venous Thrombosis/etiology , Venous Thrombosis/metabolism , Venous Thrombosis/prevention & control
9.
J Clin Endocrinol Metab ; 106(3): 622-635, 2021 03 08.
Article in English | MEDLINE | ID: covidwho-949098

ABSTRACT

This paper attempts to explain how the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus causes the complications that make coronavirus disease 2019 (COVID-19) a serious disease in specific patient subgroups. It suggests that cortisol-associated activation of the mineralocorticoid receptor (MR) in epithelial and endothelial cells infected with the virus stimulates the release of adenosine 5'-triphosphate (ATP), which then acts back on purinergic receptors. In the lung this could produce the nonproductive cough via purinergic P2X3 receptors on vagal afferent nerves. In endothelial cells it could stimulate exocytosis of Weibel-Palade bodies (WPBs) that contain angiopoietin-2, which is important in the pathogenesis of acute respiratory distress syndrome (ARDS) by increasing capillary permeability and von Willebrand factor (VWF), which mediates platelet adhesion to the endothelium and hence clotting. Angiopoietin-2 and VWF levels both are markedly elevated in COVID-19-associated ARDS. This paper offers an explanation for the sex differences in SARS-CoV-2 complications and also for why these are strongly associated with age, race, diabetes, and body mass index. It also explains why individuals with blood group A have a higher risk of severe infection than those with blood group O. Dexamethasone has been shown to be of benefit in coronavirus ARDS patients and has been thought to act as an anti-inflammatory drug. This paper suggests that a major part of its effect may be due to suppression of cortisol secretion. There is an urgent need to trial the combination of dexamethasone and an MR antagonist such as spironolactone to more effectively block the MR and hence the exocytosis of WPBs.


Subject(s)
Adenosine Triphosphate/metabolism , COVID-19/complications , Hydrocortisone/pharmacology , Receptors, Mineralocorticoid/agonists , Angiotensin-Converting Enzyme 2/physiology , Blood Coagulation Disorders/etiology , Blood Coagulation Disorders/metabolism , Blood Coagulation Disorders/virology , COVID-19/metabolism , COVID-19/pathology , Dexamethasone/therapeutic use , Eplerenone/therapeutic use , Humans , Hydrocortisone/adverse effects , Hydrocortisone/metabolism , Mineralocorticoid Receptor Antagonists/therapeutic use , Models, Biological , Paracrine Communication/drug effects , Receptors, Mineralocorticoid/metabolism , Receptors, Purinergic/physiology , Respiratory Distress Syndrome/complications , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/virology , SARS-CoV-2/drug effects , SARS-CoV-2/pathogenicity , Severity of Illness Index , Spironolactone/therapeutic use , COVID-19 Drug Treatment
10.
Blood ; 136(11): 1330-1341, 2020 09 10.
Article in English | MEDLINE | ID: covidwho-788623

ABSTRACT

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


Subject(s)
Betacoronavirus/immunology , Blood Coagulation Disorders/pathology , Blood Platelets/pathology , Coronavirus Infections/complications , Monocytes/pathology , Pneumonia, Viral/complications , Thromboplastin/metabolism , Adult , Biomarkers/metabolism , Blood Coagulation Disorders/immunology , Blood Coagulation Disorders/metabolism , Blood Coagulation Disorders/virology , Blood Platelets/metabolism , Blood Platelets/virology , COVID-19 , Case-Control Studies , Coronavirus Infections/immunology , Coronavirus Infections/metabolism , Coronavirus Infections/virology , Female , Follow-Up Studies , Humans , Male , Middle Aged , Monocytes/metabolism , Monocytes/virology , P-Selectin/metabolism , Pandemics , Platelet Activation , Pneumonia, Viral/immunology , Pneumonia, Viral/metabolism , Pneumonia, Viral/virology , Prognosis , Prospective Studies , SARS-CoV-2 , Survival Rate
11.
BMC Pregnancy Childbirth ; 20(1): 481, 2020 Aug 24.
Article in English | MEDLINE | ID: covidwho-727268

ABSTRACT

BACKGROUND: The world's understanding of COVID-19 continues to evolve as the scientific community discovers unique presentations of this disease. This case report depicts an unexpected intraoperative coagulopathy during a cesarean section in an otherwise asymptomatic patient who was later found to have COVID-19. This case suggests that there may be a higher risk for intrapartum bleeding in the pregnant, largely asymptomatic COVID-positive patient with more abnormal COVID laboratory values. CASE: The case patient displayed D-Dimer elevations beyond what is typically observed among this hospital's COVID-positive peripartum population and displayed significantly more oozing than expected intraoperatively, despite normal prothrombin time, international normalized ratio, fibrinogen, and platelets. CONCLUSION: There is little published evidence on the association between D-Dimer and coagulopathy among the pregnant population infected with SARS-CoV-2. This case report contributes to the growing body of evidence on the effects of COVID-19 in pregnancy. A clinical picture concerning for intraoperative coagulopathy may be associated with SARS-CoV-2 infection during cesarean sections, and abnormal COVID laboratory tests, particularly D-Dimer, may help identify the patients in which this presentation occurs.


Subject(s)
Blood Coagulation Disorders/blood , Blood Loss, Surgical , Breech Presentation/surgery , Cesarean Section , Coronavirus Infections/blood , Pneumonia, Viral/blood , Pregnancy Complications, Hematologic/blood , Pregnancy Complications, Infectious/blood , Adult , Antifibrinolytic Agents/therapeutic use , Betacoronavirus , Blood Coagulation Disorders/drug therapy , Blood Coagulation Disorders/metabolism , C-Reactive Protein/metabolism , COVID-19 , Cautery , Coronavirus Infections/diagnosis , Coronavirus Infections/metabolism , Female , Fibrin Fibrinogen Degradation Products/metabolism , Fibrinogen/metabolism , Hemostasis, Surgical , Humans , International Normalized Ratio , Methylergonovine/therapeutic use , Oligohydramnios , Oxytocics/therapeutic use , Oxytocin/therapeutic use , Pandemics , Platelet Count , Pneumonia, Viral/diagnosis , Pneumonia, Viral/metabolism , Pregnancy , Pregnancy Complications, Hematologic/drug therapy , Pregnancy Complications, Hematologic/metabolism , Pregnancy Complications, Infectious/diagnosis , Pregnancy Complications, Infectious/metabolism , Prothrombin Time , SARS-CoV-2 , Tranexamic Acid/therapeutic use , Uterine Inertia/drug therapy
12.
Lancet Haematol ; 7(8): e575-e582, 2020 Aug.
Article in English | MEDLINE | ID: covidwho-624336

ABSTRACT

BACKGROUND: An important feature of severe acute respiratory syndrome coronavirus 2 pathogenesis is COVID-19-associated coagulopathy, characterised by increased thrombotic and microvascular complications. Previous studies have suggested a role for endothelial cell injury in COVID-19-associated coagulopathy. To determine whether endotheliopathy is involved in COVID-19-associated coagulopathy pathogenesis, we assessed markers of endothelial cell and platelet activation in critically and non-critically ill patients admitted to the hospital with COVID-19. METHODS: In this single-centre cross-sectional study, hospitalised adult (≥18 years) patients with laboratory-confirmed COVID-19 were identified in the medical intensive care unit (ICU) or a specialised non-ICU COVID-19 floor in our hospital. Asymptomatic, non-hospitalised controls were recruited as a comparator group for biomarkers that did not have a reference range. We assessed markers of endothelial cell and platelet activation, including von Willebrand Factor (VWF) antigen, soluble thrombomodulin, soluble P-selectin, and soluble CD40 ligand, as well as coagulation factors, endogenous anticoagulants, and fibrinolytic enzymes. We compared the level of each marker in ICU patients, non-ICU patients, and controls, where applicable. We assessed correlations between these laboratory results with clinical outcomes, including hospital discharge and mortality. Kaplan-Meier analysis was used to further explore the association between biochemical markers and survival. FINDINGS: 68 patients with COVID-19 were included in the study from April 13 to April 24, 2020, including 48 ICU and 20 non-ICU patients, as well as 13 non-hospitalised, asymptomatic controls. Markers of endothelial cell and platelet activation were significantly elevated in ICU patients compared with non-ICU patients, including VWF antigen (mean 565% [SD 199] in ICU patients vs 278% [133] in non-ICU patients; p<0·0001) and soluble P-selectin (15·9 ng/mL [4·8] vs 11·2 ng/mL [3·1]; p=0·0014). VWF antigen concentrations were also elevated above the normal range in 16 (80%) of 20 non-ICU patients. We found mortality to be significantly correlated with VWF antigen (r = 0·38; p=0·0022) and soluble thrombomodulin (r = 0·38; p=0·0078) among all patients. In all patients, soluble thrombomodulin concentrations greater than 3·26 ng/mL were associated with lower rates of hospital discharge (22 [88%] of 25 patients with low concentrations vs 13 [52%] of 25 patients with high concentrations; p=0·0050) and lower likelihood of survival on Kaplan-Meier analysis (hazard ratio 5·9, 95% CI 1·9-18·4; p=0·0087). INTERPRETATION: Our findings show that endotheliopathy is present in COVID-19 and is likely to be associated with critical illness and death. Early identification of endotheliopathy and strategies to mitigate its progression might improve outcomes in COVID-19. FUNDING: This work was supported by a gift donation from Jack Levin to the Benign Hematology programme at Yale, and the National Institutes of Health.


Subject(s)
Betacoronavirus/pathogenicity , Blood Coagulation Disorders/pathology , Coronavirus Infections/complications , Endothelium, Vascular/pathology , Pneumonia, Viral/complications , Vascular Diseases/pathology , Adult , Aged , Aged, 80 and over , Biomarkers/metabolism , Blood Coagulation Disorders/etiology , Blood Coagulation Disorders/metabolism , COVID-19 , Coronavirus Infections/virology , Critical Illness , Cross-Sectional Studies , Endothelium, Vascular/metabolism , Female , Follow-Up Studies , Humans , Intensive Care Units , Male , Middle Aged , Pandemics , Pneumonia, Viral/virology , Prognosis , SARS-CoV-2 , Vascular Diseases/etiology , Vascular Diseases/metabolism , Young Adult
13.
Diagnosis (Berl) ; 7(4): 365-372, 2020 Nov 18.
Article in English | MEDLINE | ID: covidwho-615210

ABSTRACT

Coronavirus Disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a respiratory disease, which can evolve into multi-organ failure (MOF), leading to death. Several biochemical alterations have been described in COVID-19 patients. To date, many biomarkers reflecting the main pathophysiological characteristics of the disease have been identified and associated with the risk of developing severe disease. Lymphopenia represents the hallmark of the disease, and it can be detected since the early stage of infection. Increased levels of several inflammatory biomarkers, including c-reactive protein, have been found in COVID-19 patients and associated with an increased risk of severe disease, which is characterised by the so-called "cytokine storm". Also, the increase of cardiac and liver dysfunction biomarkers has been associated with poor outcome. In this review, we provide an overview of the main biochemical characteristics of COVID-19 and the associated biomarkers alterations.


Subject(s)
Betacoronavirus/genetics , Coronavirus Infections/metabolism , Pneumonia, Viral/metabolism , Aged , Aged, 80 and over , Betacoronavirus/isolation & purification , Biomarkers , Blood Coagulation Disorders/etiology , Blood Coagulation Disorders/metabolism , C-Reactive Protein/analysis , COVID-19 , Coronavirus Infections/classification , Coronavirus Infections/complications , Coronavirus Infections/epidemiology , Coronavirus Infections/virology , Cytokines/metabolism , Disease Progression , Humans , Inflammation/complications , Inflammation/metabolism , Inflammation/virology , Kidney Diseases/metabolism , Kidney Diseases/physiopathology , Liver Diseases/etiology , Liver Diseases/metabolism , Lymphopenia/etiology , Muscles/injuries , Muscles/metabolism , Myocardial Infarction/etiology , Myocardial Infarction/metabolism , Pandemics/classification , Pneumonia, Viral/classification , Pneumonia, Viral/epidemiology , Pneumonia, Viral/virology , SARS-CoV-2 , Severity of Illness Index , Water-Electrolyte Balance/physiology
14.
Ann Clin Lab Sci ; 50(3): 295-298, 2020 May.
Article in English | MEDLINE | ID: covidwho-614724

ABSTRACT

The 2019 novel coronavirus (SARS-CoV2) is the causal agent of the newly-termed Coronavirus Disease 2019 (COVID-19). In January 2020, the World Health Association (WHO) declared the CO-VID-19 as an epidemic. Abnormal coagulation parameters in COVID-19 patients currently are considered as prognostic factors of severity. Our aim is to summarize the current data available in the literature. MATERIALS AND METHODS: An electronic search was performed in the Database of publications on coronavirus disease (COVID-19) of the World Health Organization. Thrombin Time (TT), Prothrombin Time (PT), Fibrinogen (FIB), Activated Partial Thromboplastin Time (APPT), and D-Dimer have been detected as parameters to study in every COVID-19 patient. CLINICAL APPLICATION: The coagulation function panel has been described to be altered in critical COVID-2019 patients. DIC, which plays an important role in advanced stage, is known to be associated with sepsis. Anticoagulant therapy, mainly with low molecular weight heparin (LMWH), appears to be associated with better prognosis in patients with severe COV-ID-19. DISCUSSION: Coagulation function in patients with SARS-CoV2 infection is significantly deranged compared with normal patients. FIB and D-Dimer/FDP are the most significantly altered values and the early deetection of alteration could be useful to address therapies. D-Dimer/FDP (DD/FDP) alteration correlates with severity. Markedly elevated D-Dimer can be used to guide the introduction of anticoagulation therapy and evaluate prognosis of COVID-19. In every patient admitted with SARS-CoV2 infection PT, FIB, D-Dimer/FDP, and platelets must be ordered. We suggest daily extraction for every patient admitted and tested positive for COVID-19.


Subject(s)
Betacoronavirus/isolation & purification , Biomarkers/analysis , Blood Coagulation Disorders/diagnosis , Coronavirus Infections/complications , Pneumonia, Viral/complications , Blood Coagulation Disorders/etiology , Blood Coagulation Disorders/metabolism , Blood Coagulation Tests , COVID-19 , Coronavirus Infections/virology , Humans , Pandemics , Pneumonia, Viral/virology , Prognosis , SARS-CoV-2
15.
Emerg Microbes Infect ; 9(1): 1514-1522, 2020 Dec.
Article in English | MEDLINE | ID: covidwho-611844

ABSTRACT

We previously made the hypothesis that STING contributes to COVID-19. The present review detail new arguments for over-activation of STING pathways in COVID-19, following the description of hyper-coagulability and Kawasaki-like diseases in children. Indeed, Kawasaki disease is induced by overreaction of innate cells following exposition to various viruses, including herpes viruses which trigger STING. It predisposes to diffuse vasculitis and aneurysms, whereas STING is over-expressed in arterial aneurisms. The redness at the inoculation site of bacillus Calmette-Guérin, a specific feature of Kawasaki disease, is reproduced by activation of the STING pathway, which is inhibited upstream by aspirin, intravenous immunoglobulins, and Vitamin-D. SARS-CoV2 binding to ACE2 can lead to excessive angiotensin II signaling, which activates the STING pathway in mice. Over-activation of the STING-pathway promotes hyper-coagulability through release of interferon-ß and tissue factor by monocytes-macrophages. Aspirin and dipyridamole, besides their anti-platelet activity, also reduce tissue factor procoagulant activity, and aspirin inhibits the STING pathway upstream of STING. Aspirin and dipyridamole may be used, in combination with drugs blocking downstream the activation of the STING pathway, like inhibitors of IL-6R and JAK/STAT pathways. The risk of bleeding should be low as bleeding has not been reported in severe COVID-19 patients.


Subject(s)
Coronavirus Infections/complications , Membrane Proteins/metabolism , Mucocutaneous Lymph Node Syndrome/etiology , Pneumonia, Viral/complications , Angiotensin II/metabolism , Animals , Aspirin/therapeutic use , Blood Coagulation Disorders/drug therapy , Blood Coagulation Disorders/metabolism , Blood Coagulation Disorders/virology , COVID-19 , Coronavirus Infections/drug therapy , Coronavirus Infections/metabolism , Dipyridamole/therapeutic use , Humans , Immunoglobulins, Intravenous/therapeutic use , Interferons/metabolism , Mice , Mucocutaneous Lymph Node Syndrome/metabolism , Pandemics , Platelet Aggregation Inhibitors/therapeutic use , Pneumonia, Viral/drug therapy , Pneumonia, Viral/metabolism , Signal Transduction , Thrombosis/drug therapy , Thrombosis/metabolism , Thrombosis/virology
16.
Blood ; 136(11): 1317-1329, 2020 09 10.
Article in English | MEDLINE | ID: covidwho-612131

ABSTRACT

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


Subject(s)
Betacoronavirus/isolation & purification , Blood Coagulation Disorders/pathology , Blood Platelets/pathology , Coronavirus Infections/complications , Pneumonia, Viral/complications , Transcriptome , Biomarkers , Blood Coagulation Disorders/genetics , Blood Coagulation Disorders/metabolism , Blood Coagulation Disorders/virology , Blood Platelets/metabolism , Blood Platelets/virology , COVID-19 , Case-Control Studies , Coronavirus Infections/genetics , Coronavirus Infections/metabolism , Coronavirus Infections/virology , Female , Follow-Up Studies , Gene Expression Profiling , Humans , Male , Middle Aged , Pandemics , Pneumonia, Viral/genetics , Pneumonia, Viral/metabolism , Pneumonia, Viral/virology , Prognosis , Prospective Studies , SARS-CoV-2
SELECTION OF CITATIONS
SEARCH DETAIL