Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
PLoS Genet ; 18(3): e1010042, 2022 03.
Article in English | MEDLINE | ID: covidwho-1793655

ABSTRACT

In November 2021, the COVID-19 pandemic death toll surpassed five million individuals. We applied Mendelian randomization including >3,000 blood proteins as exposures to identify potential biomarkers that may indicate risk for hospitalization or need for respiratory support or death due to COVID-19, respectively. After multiple testing correction, using genetic instruments and under the assumptions of Mendelian Randomization, our results were consistent with higher blood levels of five proteins GCNT4, CD207, RAB14, C1GALT1C1, and ABO being causally associated with an increased risk of hospitalization or respiratory support/death due to COVID-19 (ORs = 1.12-1.35). Higher levels of FAAH2 were solely associated with an increased risk of hospitalization (OR = 1.19). On the contrary, higher levels of SELL, SELE, and PECAM-1 decrease risk of hospitalization or need for respiratory support/death (ORs = 0.80-0.91). Higher levels of LCTL, SFTPD, KEL, and ATP2A3 were solely associated with a decreased risk of hospitalization (ORs = 0.86-0.93), whilst higher levels of ICAM-1 were solely associated with a decreased risk of respiratory support/death of COVID-19 (OR = 0.84). Our findings implicate blood group markers and binding proteins in both hospitalization and need for respiratory support/death. They, additionally, suggest that higher levels of endocannabinoid enzymes may increase the risk of hospitalization. Our research replicates findings of blood markers previously associated with COVID-19 and prioritises additional blood markers for risk prediction of severe forms of COVID-19. Furthermore, we pinpoint druggable targets potentially implicated in disease pathology.


Subject(s)
Blood Proteins/metabolism , COVID-19/blood , COVID-19/pathology , Biomarkers/analysis , Biomarkers/blood , Blood Proteins/analysis , Blood Proteins/genetics , COVID-19/diagnosis , COVID-19/mortality , Causality , Genome-Wide Association Study , Hospitalization , Humans , Mendelian Randomization Analysis , Mortality , Pandemics , Polymorphism, Single Nucleotide , Prognosis , Proteome/analysis , Proteome/genetics , Proteome/metabolism , Respiratory Insufficiency/blood , Respiratory Insufficiency/diagnosis , Respiratory Insufficiency/mortality , Respiratory Insufficiency/pathology , Risk Factors , SARS-CoV-2/physiology , Severity of Illness Index
2.
Nat Commun ; 13(1): 946, 2022 02 17.
Article in English | MEDLINE | ID: covidwho-1709499

ABSTRACT

COVID-19 complications still present a huge burden on healthcare systems and warrant predictive risk models to triage patients and inform early intervention. Here, we profile 893 plasma proteins from 50 severe and 50 mild-moderate COVID-19 patients, and 50 healthy controls, and show that 375 proteins are differentially expressed in the plasma of severe COVID-19 patients. These differentially expressed plasma proteins are implicated in the pathogenesis of COVID-19 and present targets for candidate drugs to prevent or treat severe complications. Based on the plasma proteomics and clinical lab tests, we also report a 12-plasma protein signature and a model of seven routine clinical tests that validate in an independent cohort as early risk predictors of COVID-19 severity and patient survival. The risk predictors and candidate drugs described in our study can be used and developed for personalized management of SARS-CoV-2 infected patients.


Subject(s)
Blood Proteins/analysis , COVID-19/mortality , COVID-19/pathology , Severity of Illness Index , Adult , Cytokines/blood , Female , Humans , Male , Middle Aged , Prognosis , Proteomics/methods , SARS-CoV-2/drug effects , Young Adult , COVID-19 Drug Treatment
3.
Blood Coagul Fibrinolysis ; 32(8): 550-555, 2021 Dec 01.
Article in English | MEDLINE | ID: covidwho-1526212

ABSTRACT

Coronavirus-related disease-2019 (COVID-19)-associated coagulopathy presents predominantly with thrombosis and leads to complications in close association with inflammatory process. Soluble endothelial protein C receptor (sEPCR), which is the soluble form of EPCR, reduces the anticoagulant and anti-inflammatory activity of activated protein C. The purpose of this study is to investigate the relationship between sEPCR and the laboratory parameters and thorax computed tomography (CT) findings in the course of COVID-19. Twenty-five laboratory-confirmed [reverse transcription-quantitative polimerase chain reaction (RT-qPCR) positive] and 24 clinically diagnosed (RT-qPCR negative) COVID-19 patients were enrolled in the study. Blood specimens were collected for sEPCR and haematological and biochemical parameter measurement. Thorax CT was performed to detect COVID-19 findings. These parameters from RT-qPCR positive and negative patients were then compared. Although there was no difference between the groups in terms of symptoms, the time between the onset of symptoms and the admission time was shorter in RT-qPCR positive group (P = 0.000). sEPCR levels were significantly higher in the RT-qPCR positive group (P = 0.011). Patients with ground-glass opacity and bilateral involvement on thorax CT have higher serum sEPCR levels (P = 0.012 and 0.043, respectively). This study has shown for the first time that serum sEPCR levels, which is a member of coagulation cascade and has also been reported to be associated with inflammation, is higher in patients with positive RT-qPCR test and patients with GGO or bilateral involvement on thorax CT regardless of the PCR result.


Subject(s)
COVID-19/blood , Endothelial Protein C Receptor/blood , SARS-CoV-2 , Thrombophilia/blood , Adult , Aged , Aged, 80 and over , Biomarkers , Blood Glucose/analysis , Blood Proteins/analysis , COVID-19/complications , COVID-19/diagnostic imaging , Female , Humans , Lung/diagnostic imaging , Male , Middle Aged , Solubility , Thrombophilia/etiology , Tomography, X-Ray Computed
4.
Mol Cell Proteomics ; 20: 100159, 2021.
Article in English | MEDLINE | ID: covidwho-1525879

ABSTRACT

Viruses hijack host metabolic pathways for their replicative advantage. In this study, using patient-derived multiomics data and in vitro infection assays, we aimed to understand the role of key metabolic pathways that can regulate severe acute respiratory syndrome coronavirus-2 reproduction and their association with disease severity. We used multiomics platforms (targeted and untargeted proteomics and untargeted metabolomics) on patient samples and cell-line models along with immune phenotyping of metabolite transporters in patient blood cells to understand viral-induced metabolic modulations. We also modulated key metabolic pathways that were identified using multiomics data to regulate the viral reproduction in vitro. Coronavirus disease 2019 disease severity was characterized by increased plasma glucose and mannose levels. Immune phenotyping identified altered expression patterns of carbohydrate transporter, glucose transporter 1, in CD8+ T cells, intermediate and nonclassical monocytes, and amino acid transporter, xCT, in classical, intermediate, and nonclassical monocytes. In in vitro lung epithelial cell (Calu-3) infection model, we found that glycolysis and glutaminolysis are essential for virus replication, and blocking these metabolic pathways caused significant reduction in virus production. Taken together, we therefore hypothesized that severe acute respiratory syndrome coronavirus-2 utilizes and rewires pathways governing central carbon metabolism leading to the efflux of toxic metabolites and associated with disease severity. Thus, the host metabolic perturbation could be an attractive strategy to limit the viral replication and disease severity.


Subject(s)
Blood Proteins/metabolism , COVID-19/etiology , SARS-CoV-2/physiology , Adult , Aged , Amino Acid Transport System y+/blood , Amino Acids/blood , Biomarkers/blood , Blood Proteins/analysis , COVID-19/metabolism , COVID-19/virology , Carbohydrates/blood , Case-Control Studies , Glucose Transporter Type 1/blood , Hospitalization , Humans , Immunophenotyping , Mannose/blood , Mannose-Binding Lectin/blood , Middle Aged , Severity of Illness Index , Virus Replication
5.
Front Immunol ; 12: 738093, 2021.
Article in English | MEDLINE | ID: covidwho-1518484

ABSTRACT

Disease caused by SARS-CoV-2 coronavirus (COVID-19) led to significant morbidity and mortality worldwide. A systemic hyper-inflammation characterizes severe COVID-19 disease, often associated with acute respiratory distress syndrome (ARDS). Blood biomarkers capable of risk stratification are of great importance in effective triage and critical care of severe COVID-19 patients. Flow cytometry and next-generation sequencing were done on peripheral blood cells and urokinase-type plasminogen activator receptor (suPAR), and cytokines were measured from and mass spectrometry-based proteomics was done on plasma samples from an Indian cohort of COVID-19 patients. Publicly available single-cell RNA sequencing data were analyzed for validation of primary data. Statistical analyses were performed to validate risk stratification. We report here higher plasma abundance of suPAR, expressed by an abnormally expanded myeloid cell population, in severe COVID-19 patients with ARDS. The plasma suPAR level was found to be linked to a characteristic plasma proteome, associated with coagulation disorders and complement activation. Receiver operator characteristic curve analysis to predict mortality identified a cutoff value of suPAR at 1,996.809 pg/ml (odds ratio: 2.9286, 95% confidence interval 1.0427-8.2257). Lower-than-cutoff suPAR levels were associated with a differential expression of the immune transcriptome as well as favorable clinical outcomes, in terms of both survival benefit (hazard ratio: 0.3615, 95% confidence interval 0.1433-0.912) and faster disease remission in our patient cohort. Thus, we identified suPAR as a key pathogenic circulating molecule linking systemic hyperinflammation to the hypercoagulable state and stratifying clinical outcomes in severe COVID-19 patients with ARDS.


Subject(s)
COVID-19/blood , Receptors, Urokinase Plasminogen Activator/blood , SARS-CoV-2 , Adult , Aged , Blood Coagulation Disorders/blood , Blood Coagulation Disorders/immunology , Blood Proteins/analysis , COVID-19/immunology , Cytokines/blood , Humans , Inflammation/blood , Inflammation/immunology , Middle Aged , Myeloid Cells/immunology , Proteome/analysis , Randomized Controlled Trials as Topic , Respiratory Distress Syndrome/blood , Respiratory Distress Syndrome/immunology , Severity of Illness Index , Young Adult
6.
J Mol Med (Berl) ; 100(2): 285-301, 2022 02.
Article in English | MEDLINE | ID: covidwho-1505851

ABSTRACT

The risk of severe COVID-19 increases with age as older patients are at highest risk. Thus, there is an urgent need to identify how severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) interacts with blood components during aging. We investigated the whole blood transcriptome from the Genotype-Tissue Expression (GTEx) database to explore differentially expressed genes (DEGs) translated into proteins interacting with viral proteins during aging. From 22 DEGs in aged blood, FASLG, CTSW, CTSE, VCAM1, and BAG3 were associated with immune response, inflammation, cell component and adhesion, and platelet activation/aggregation. Males and females older than 50 years old overexpress FASLG, possibly inducing a hyperinflammatory cascade. The expression of cathepsins (CTSW and CTSE) and the anti-apoptotic co-chaperone molecule BAG3 also increased throughout aging in both genders. By exploring single-cell RNA-sequencing data from peripheral blood of SARS-CoV-2-infected patients, we found FASLG and CTSW expressed in natural killer cells and CD8 + T lymphocytes, whereas BAG3 was expressed mainly in CD4 + T cells, naive T cells, and CD14 + monocytes. In addition, T cell exhaustion was associated with increased expression of CCL4L2 and DUSP4 over blood aging. LAG3, PDCD1, TIGIT, VCAM1, HLA-DRA, and TOX also increased in individuals aged 60-69 years old; conversely, the RGS2 gene decreased with aging. We further identified a distinct gene expression profile associated with type I interferon signaling following blood aging. These results revealed changes in blood molecules potentially related to SARS-CoV-2 infection throughout aging, emphasizing them as therapeutic candidates for aggressive clinical manifestation of COVID-19. KEY MESSAGES: • Prediction of host-viral interactions in the whole blood transcriptome during aging. • Expression levels of FASLG, CTSW, CTSE, VCAM1, and BAG3 increase in aged blood. • Blood interactome reveals targets involved with immune response, inflammation, and blood clots. • SARS-CoV-2-infected patients with high viral load showed FASLG overexpression. • Gene expression profile associated with T cell exhaustion and type I interferon signaling were affected with blood aging.


Subject(s)
Aging/blood , Blood Proteins/analysis , COVID-19/genetics , SARS-CoV-2/pathogenicity , Transcriptome , Adult , Aged , Aging/genetics , Blood/metabolism , Blood Chemical Analysis , Blood Proteins/genetics , Blood Proteins/metabolism , Blood Vessels/metabolism , Blood Vessels/virology , COVID-19/blood , COVID-19/immunology , COVID-19/physiopathology , Cardiovascular Physiological Phenomena/genetics , Cardiovascular System/metabolism , Cardiovascular System/virology , Cohort Studies , Female , Genetic Association Studies , Humans , Immunity, Innate/genetics , Male , Middle Aged , Young Adult
7.
Int J Lab Hematol ; 43(6): 1302-1308, 2021 Dec.
Article in English | MEDLINE | ID: covidwho-1288297

ABSTRACT

INTRODUCTION: We aimed to identify the associations between the lymphocytes (LYM) absolute count on admission and clinical outcomes in COVID-19 patients. METHODS: In this retrospective study, 224 COVID-19 patients who were admitted to General Hospital of Central Theater Command of the PLA from January 22 to April 4, 2020, were consecutively included. These patients were divided into the lymphopenia group and the nonlymphopenia group according to whether the LYM count on admission was below the normal range. RESULTS: During hospitalization, patients in the lymphopenia group have a much higher all-cause mortality (14.5% vs 0.0%; P < .001) and an evidently longer length of hospital stay (24.0 vs 17.5 days; P < .001) than patients in the nonlymphopenia group. The correlation analysis results indicated that the LYM count was negatively correlated with the values of NEU (R = -.2886, P < .001), PT (R = -.2312, P < .001), FIB (R = -.2954, P < .001), D-D (R = -.3554, P < .001), CRP (R = -.4899, P < .001), IL-6 (R = -.5459, P < .001), AST (R = -.2044, P < .01), Cr (R = -.1350, P < .05), CPK (R = -.2119, P < .01), CK-Mb (R = -.1760, P < .01), and LDH (R = -.4330, P < .001), and was positively correlated with the count of PLT (R = .2679, P < .001). In addition, LYM as a continuous variable was associated with 97% decreased risk of in-hospital mortality in the fully adjusted models (OR = 0.03, 95%CI, 0.00-0.37, P < .001). DISCUSSION: LYM screening on admission is a critical predictor for assessment of disease severity and clinical outcomes in patients with COVID-19, and lymphopenia substantially correlates with poor clinical outcomes.


Subject(s)
COVID-19/blood , Lymphocyte Count , SARS-CoV-2 , Adult , Aged , Biomarkers/blood , Blood Cell Count , Blood Coagulation Tests , Blood Proteins/analysis , COVID-19/mortality , China/epidemiology , Creatinine/blood , Female , Hospital Mortality , Hospitals, General/statistics & numerical data , Humans , Lymphopenia/blood , Lymphopenia/etiology , Male , Mass Screening , Middle Aged , Patient Admission , Prognosis , Retrospective Studies , Severity of Illness Index , Treatment Outcome
8.
Clin Appl Thromb Hemost ; 27: 10760296211027653, 2021.
Article in English | MEDLINE | ID: covidwho-1286792

ABSTRACT

Identifying a hypercoagulable state in patients with COVID-19 may help identify those at risk for virus-induced thromboembolic events and improve clinical outcomes using personalized therapeutic approaches. Herein, we aimed to perform a global assessment of the patients' hemostatic system with COVID-19 using rotational thromboelastometry (ROTEM) and to describe whether patients with different disease severities present different coagulation profiles. Together with 37 healthy volunteers, a total of 65 patients were included and then classified as having mild, moderate, and severe disease depending on clinical severity. Peripheral blood samples were collected and analyzed using a ROTEM Coagulation Analyzer. Also, complete blood count and coagulation parameters including prothrombin time, activated partial thromboplastin time, fibrinogen levels, and D-dimer levels were measured at admission. EXTEM and INTEM MCF (P < 0.001) values were significantly higher and the EXTEM CFT (P = 0.002) value was significantly lower in patients with COVID-19 when compared with controls. In particular, patients with the severe disease showed a significant decrease in CFT (P < 0.001) and an increase in MCF (P < 0.001) in both INTEM and EXTEM assays compared with patients with the non-severe disease. Correlation analysis revealed significant correlations between ROTEM parameters and other coagulation parameters. There were significant positive correlations between fibrinogen, D-dimer, platelet count, and MCF in both EXTEM and INTEM assays. Our data demonstrate thromboelastographic signs of hypercoagulability in patients with COVID-19, which is more pronounced in patients with increased disease severity. Therefore, ROTEM analysis can classify subsets of patients with COVID-19 at significant thrombotic risk and assist in clinical decisions.


Subject(s)
COVID-19/blood , SARS-CoV-2 , Thrombelastography , Thrombophilia/etiology , Adult , Blood Cell Count , Blood Coagulation Tests , Blood Proteins/analysis , Case-Control Studies , Female , Humans , Male , Middle Aged , Prospective Studies , Severity of Illness Index , Thrombophilia/blood , Thrombophilia/diagnosis
9.
Mol Cell Proteomics ; 20: 100113, 2021.
Article in English | MEDLINE | ID: covidwho-1275575

ABSTRACT

Severe coronavirus disease 2019 (COVID-19) can result in pneumonia and acute respiratory failure. Accumulation of mucus in the airways is a hallmark of the disease and can result in hypoxemia. Here, we show that quantitative proteome analysis of the sputum from severe patients with COVID-19 reveal high levels of neutrophil extracellular trap (NET) components, which was confirmed by microscopy. Extracellular DNA from excessive NET formation can increase sputum viscosity and lead to acute respiratory distress syndrome. Recombinant human DNase (Pulmozyme; Roche) has been shown to be beneficial in reducing sputum viscosity and improve lung function. We treated five patients pwith COVID-19 resenting acute symptoms with clinically approved aerosolized Pulmozyme. No adverse reactions to the drug were seen, and improved oxygen saturation and recovery in all severely ill patients with COVID-19 was observed after therapy. Immunofluorescence and proteome analysis of sputum and blood plasma samples after treatment revealed a marked reduction of NETs and a set of statistically significant proteome changes that indicate reduction of hemorrhage, plasma leakage and inflammation in the airways, and reduced systemic inflammatory state in the blood plasma of patients. Taken together, the results indicate that NETs contribute to acute respiratory failure in COVID-19 and that degrading NETs may reduce dependency on external high-flow oxygen therapy in patients. Targeting NETs using recombinant human DNase may have significant therapeutic implications in COVID-19 disease and warrants further studies.


Subject(s)
COVID-19 Drug Treatment , Deoxyribonuclease I/pharmacology , Extracellular Traps/metabolism , Proteome/analysis , Aged , Blood Proteins/analysis , COVID-19/metabolism , COVID-19/therapy , Female , Fluorescent Antibody Technique , Humans , Male , Middle Aged , Recombinant Proteins/pharmacology , Severity of Illness Index , Sputum/drug effects , Sputum/metabolism , Sputum/virology , Systemic Inflammatory Response Syndrome/drug therapy , Systemic Inflammatory Response Syndrome/metabolism , Systemic Inflammatory Response Syndrome/virology
10.
Int J Lab Hematol ; 43(6): 1291-1301, 2021 Dec.
Article in English | MEDLINE | ID: covidwho-1258939

ABSTRACT

INTRODUCTION: The clinical and laboratory features of severe COVID-19 infection overlap with those of hemophagocytic lymphohistiocytosis (HLH), a hyperinflammatory disorder often associated with several viral infections. The clinical syndrome of HLH encompasses fever, organomegaly, cytopenias, hyperferritinemia, hypertriglyceridemia, raised transaminases, hypofibrinogenemia, absent natural killer (NK) cell activity, increased soluble CD25 and hemophagocytic lymphohistiocytosis in bone marrow, spleen, and lymph nodes. METHODS: We analyzed clinicopathological and laboratory features of thirteen patients with severe COVID-19 infection suspected to have HLH and found to have hemophagocytic histiocytosis on bone marrow examination (BME). RESULTS: Five of thirteen (38.46%) patients fulfilled five of eight HLH 2004 criteria and/or had a H-score ≥169. Three (23.08%) satisfied four of eight and remainder five (38.46%) satisfied three of eight HLH 2004 criteria. Fever, raised serum ferritin (13/13, 100%), transaminases (9/13, 69.23%), triglycerides (4/13, 30.76%), cytopenias (5/13, 38.46%), hypofibrinogenemia (2/13, 15.38%), and organomegaly (1/13, 7.69%) were observed in our patients. BME showed hemophagocytic histiocytosis without lymphocytosis in all. Contrary to HLH, lymphocytopenia (11/13, 84.61%), leukocytosis (7/13, 53.84%), neutrophilia (7/13, 53.84%), and hyperfibrinogenemia (7/13, 53.84%) were observed. Serum CRP, LDH, and plasma D-dimer were elevated in all, while serum albumin was decreased in 12 of 13 (92.3%) patients. Five patients recovered with high-dose pulsed corticosteroid therapy. CONCLUSION: The immune response associated with severe COVID-19 infection is similar to HLH with few differences. HLH should be suspected in severe COVID-19 infection although all patients may not fulfill required HLH diagnostic criteria. BME should be done in suspected cases so that appropriate therapy may be initiated early.


Subject(s)
Bone Marrow/pathology , COVID-19/complications , Lymphohistiocytosis, Hemophagocytic/etiology , SARS-CoV-2 , Adrenal Cortex Hormones/therapeutic use , Adult , Aged , Biomarkers/blood , Blood Proteins/analysis , Bone Marrow Examination , COVID-19/immunology , Creatinine/blood , Diagnosis, Differential , Female , Humans , Leukocyte Count , Lymphohistiocytosis, Hemophagocytic/blood , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/pathology , Male , Middle Aged , Neutrophils , Severity of Illness Index , Symptom Assessment , Triglycerides/blood
11.
Front Endocrinol (Lausanne) ; 12: 651009, 2021.
Article in English | MEDLINE | ID: covidwho-1190304

ABSTRACT

Introduction: Patients with severe COVID-19 infections have coagulation abnormalities indicative of a hypercoagulable state, with thromboembolic complications and increased mortality. Platelets are recognized as mediators of inflammation, releasing proinflammatory and prothrombotic factors, and are hyperactivated in COVID-19 infected patients. Activated platelets have also been reported in type 2 diabetes (T2D) patients, putting these patients at higher risk for thromboembolic complications of COVID-19 infection. Methods: A case-control study of T2D (n=33) and control subjects (n=30) who underwent a hyperinsulinemic clamp to induce normoglycemia in T2D subjects: T2D: baseline glucose 7.5 ± 0.3mmol/l (135.1 ± 5.4mg/dl), reduced to 4.5 ± 0.07mmol/l (81 ± 1.2mg/dl) with 1-hour clamp; Controls: maintained at 5.1 ± 0.1mmol/l (91.9 ± 1.8mg/dl). Slow Off-rate Modified Aptamer (SOMA)-scan plasma protein measurement was used to determine a panel of platelet proteins. Results: Prothrombotic platelet proteins were elevated in T2D versus controls: platelet factor 4 (PF4, p<0.05); platelet glycoprotein VI (PGVI p<0.05); P-selectin (p<0.01) and plasminogen activator inhibitor I (PAI-1, p<0.01). In addition, the antithrombotic platelet-related proteins, plasmin (p<0.05) and heparin cofactor II (HCFII, p<0.05), were increased in T2D. Normalization of glucose in the T2D cohort had no effect on platelet protein levels. Conclusion: T2D patients have platelet hyperactivation, placing them at higher risk for thromboembolic events. When infected with COVID-19, this risk may be compounded, and their propensity for a more severe COVID-19 disease course increased. Clinical Trial Registration: https://clinicaltrials.gov/ct2/show/NCT03102801, identifier NCT03102801.


Subject(s)
Blood Platelet Disorders/blood , Blood Platelet Disorders/etiology , Blood Platelets/chemistry , Blood Proteins/analysis , COVID-19/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Hypoglycemia/blood , Hypoglycemia/complications , Aged , COVID-19/complications , Case-Control Studies , Female , Glucose Clamp Technique , Humans , Lipids/blood , Male , Middle Aged , Platelet Activation , Thromboembolism/blood , Thromboembolism/etiology
12.
Ann Palliat Med ; 10(1): 672-680, 2021 Jan.
Article in English | MEDLINE | ID: covidwho-1136693

ABSTRACT

BACKGROUND: The novel 2019 coronavirus (COVID-19) has largely abated in China; however, sporadic or imported cases are still a concern, while in other countries, the COVID-19 pandemic persists as a major health crisis. METHODS: All patients enrolled in this study were diagnosed with COVID-19 from February 21, 2020 to April 14, 2020 in Wuhan. We retrospectively analyzed the patients admitted to the ICU (137 patients) and general wards (114 patients) of Wuhan Leishenshan Hospital in China. The population characteristics, symptoms, and laboratory examination results between the patients in the ICU and those in the general wards were compared. Furthermore, the differences between the deceased patients in the ICU and those discharged from the ICU were compared. RESULTS: There were significant differences between the two groups in terms of symptoms, including fever, shortness of breath, no presence of complications, presence of 1 complication, and presence of 3 or more complications (P<0.05). There were also significant differences between the patients in terms of the laboratory examination results including elevated urea nitrogen, creatinine, direct bilirubin, aspartate aminotransferase, total protein, albumin, creatine kinase, lactate dehydrogenase, procalcitonin, erythrocyte sedimentation rate, white blood cells, C-reactive protein, prothrombin time, activated partial thromboplastin time, fibrinogen, D-dimer, interleukin 6, interleukin 8, interleukin 10, interleukin 2 receptor, tumor necrosis factor-α, troponin I, phosphokinase isoenzyme-MB, and B-type natriuretic peptide; and decreased platelets, lymphocyte absolute value, and eosinophil absolute value (<0.05). There were 45 patients who died in ICU and 57 improved and discharged patients. There were significant differences between the two groups in the number of patients that had 1 complication and 3 or more complications (P<0.05). There were also significant differences in the laboratory examination results between the patients including elevated urea nitrogen, total bilirubin, direct bilirubin, aspartate aminotransferase, procalcitonin, white blood cells, interleukin 8, interleukin 10, phosphokinase isoenzyme-MB, and B-type natriuretic peptide; and decreased platelets and eosinophil absolute value (P<0.05). CONCLUSIONS: Our findings highlight that the identified determinants may help to improve treatment of COVID-19 patients, to predict the risk of developing severe illness and to optimizing arrangement of health resources.


Subject(s)
COVID-19/blood , COVID-19/mortality , Hospitalization , Intensive Care Units , Adolescent , Adult , Aged , Aged, 80 and over , Aspartate Aminotransferases/blood , Bilirubin/blood , Blood Cell Count , Blood Coagulation Tests , Blood Proteins/analysis , Blood Sedimentation , Blood Urea Nitrogen , Creatine Kinase/blood , Creatinine/analysis , Cytokines/blood , Female , Fever/virology , Humans , L-Lactate Dehydrogenase/blood , Male , Middle Aged , Natriuretic Peptide, Brain/blood , Procalcitonin/blood , Retrospective Studies , Young Adult
13.
Front Endocrinol (Lausanne) ; 12: 638621, 2021.
Article in English | MEDLINE | ID: covidwho-1133905

ABSTRACT

Background: Women with polycystic ovary syndrome (PCOS) often have vitamin D deficiency, a known risk factor for severe COVID-19 disease. Alveolar macrophage-derived cytokines contribute to the inflammation underlying pulmonary disease in COVID-19. We sought to determine if basal macrophage activation, as a risk factor for COVID-19 infection, was present in PCOS and, if so, was further enhanced by vitamin D deficiency. Methods: A cross-sectional study in 99 PCOS and 68 control women who presented sequentially. Plasma levels of a macrophage-derived cytokine panel were determined by Slow Off-rate Modified Aptamer (SOMA)-scan plasma protein measurement. Vitamin D was measured by tandem mass spectroscopy. Results: Vitamin D was lower in PCOS women (p<0.0001) and correlated negatively with body mass index (BMI) in PCOS (r=0.28, p=0.0046). Basal macrophage activation markers CXCL5, CD163 and MMP9 were elevated, whilst protective CD200 was decreased (p<0.05); changes in these variables were related to, and fully accounted for, by BMI. PCOS and control women were then stratified according to vitamin D concentration. Vitamin D deficiency was associated with decreased CD80 and IFN-γ in PCOS and IL-12 in both groups (p<0.05). These factors, important in initiating and maintaining the immune response, were again accounted for by BMI. Conclusion: Basal macrophage activation was higher in PCOS with macrophage changes related with increased infection risk associating with vitamin D; all changes were BMI dependent, suggesting that obese PCOS with vitamin D deficiency may be at greater risk of more severe COVID-19 infection, but that it is obesity-related rather than an independent PCOS factor.


Subject(s)
COVID-19/epidemiology , Cytokines/metabolism , Macrophages/metabolism , Polycystic Ovary Syndrome/epidemiology , Vitamin D Deficiency/epidemiology , Adult , Biomarkers/analysis , Blood Proteins/analysis , Body Mass Index , COVID-19/complications , COVID-19/immunology , Cross-Sectional Studies , Female , Humans , Macrophage Activation , Macrophages/chemistry , Polycystic Ovary Syndrome/complications , Polycystic Ovary Syndrome/immunology , Risk Assessment , Tandem Mass Spectrometry , Vitamin D/blood , Vitamin D Deficiency/complications
14.
J Infect Dis ; 223(1): 19-22, 2021 01 04.
Article in English | MEDLINE | ID: covidwho-1117042

ABSTRACT

It is critical to identify potential causal targets for SARS-CoV-2, which may guide drug repurposing options. We assessed the associations between genetically predicted protein levels and COVID-19 severity. Leveraging data from the COVID-19 Host Genetics Initiative comparing 6492 hospitalized COVID-19 patients and 1 012 809 controls, we identified 18 proteins with genetically predicted levels to be associated with COVID-19 severity at a false discovery rate of <0.05, including 12 that showed an association even after Bonferroni correction. Of the 18 proteins, 6 showed positive associations and 12 showed inverse associations. In conclusion, we identified 18 candidate proteins for COVID-19 severity.


Subject(s)
Blood Proteins/analysis , COVID-19/diagnosis , COVID-19/genetics , Genetic Association Studies , Case-Control Studies , Humans , Severity of Illness Index
16.
Blood Coagul Fibrinolysis ; 32(4): 290-293, 2021 Jun 01.
Article in English | MEDLINE | ID: covidwho-1066463

ABSTRACT

Respiratory failure in coronavirus disease 2019 (COVID-19) patients is one of the most frequent causes for referral to the ICU. A significant percentage of these patients does not survive the infection due to thromboembolic complications. Furthermore, the vascular system seems also to be involved in the pathogenesis. To investigate the role of hemostasis and endothelium on the outcome of COVID-19 patients admitted to the ICU. Blood was drawn from 16 ICU COVID-19 patients for hemostatic analysis. Patients were followed-up till discharge (n = 11) or death (n = 5). Parameters related to both coagulation and fibrinolysis, though disturbed, were not associated with mortality. Contrarily, activated Von Willebrand factor was increased and ADAMTS13 levels were decreased by two-fold in nonsurvivors compared with survivors. Our data established the involvement of the Von Willebrand factor-ADAMTS13 axis in the COVID-19 pathogenesis, thereby demonstrating that these plasma proteins seem to be strong predictors for ICU mortality.


Subject(s)
ADAMTS13 Protein/blood , COVID-19/blood , Endothelium, Vascular/physiopathology , SARS-CoV-2 , von Willebrand Factor/analysis , ADAMTS13 Protein/deficiency , Aged , Aged, 80 and over , Biomarkers , Blood Proteins/analysis , COVID-19/complications , COVID-19/mortality , Cross-Sectional Studies , Endothelium, Vascular/metabolism , Extracorporeal Circulation , Female , Fibrinolysin/biosynthesis , Fibrinolysis , Hemostasis , Heparin/therapeutic use , Humans , Intensive Care Units , Male , Middle Aged , Partial Thromboplastin Time , Prognosis , SARS-CoV-2/isolation & purification , Thrombin/biosynthesis
18.
Sci Rep ; 10(1): 22418, 2020 12 29.
Article in English | MEDLINE | ID: covidwho-997951

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has infected over forty million patients worldwide. Although most coronavirus disease 2019 (COVID-19) patients have a good prognosis, some develop severe illness. Markers that define disease severity or predict clinical outcome need to be urgently developed as the mortality rate in critical cases is approximately 61.5%. In the present study, we performed in-depth proteome profiling of undepleted plasma from eight COVID-19 patients. Quantitative proteomic analysis using the BoxCar method revealed that 91 out of 1222 quantified proteins were differentially expressed depending on the severity of COVID-19. Importantly, we found 76 proteins, previously not reported, which could be novel prognostic biomarker candidates. Our plasma proteome signatures captured the host response to SARS-CoV-2 infection, thereby highlighting the role of neutrophil activation, complement activation, platelet function, and T cell suppression as well as proinflammatory factors upstream and downstream of interleukin-6, interleukin-1B, and tumor necrosis factor. Consequently, this study supports the development of blood biomarkers and potential therapeutic targets to aid clinical decision-making and subsequently improve prognosis of COVID-19.


Subject(s)
Blood Proteins/analysis , COVID-19/blood , Severity of Illness Index , Adult , Aged , Biomarkers/blood , COVID-19/mortality , COVID-19/pathology , Chromatography, High Pressure Liquid , Complement Activation/immunology , Cytokines/blood , Gene Expression Profiling , Humans , Mass Spectrometry , Middle Aged , Neutrophil Activation/immunology , Platelet Activation/immunology , Proteome/metabolism , SARS-CoV-2 , Suppressor Factors, Immunologic/blood , T-Lymphocytes/immunology
19.
Medicine (Baltimore) ; 99(45): e22971, 2020 Nov 06.
Article in English | MEDLINE | ID: covidwho-990916

ABSTRACT

Coronavirus disease 2019 (COVID-19) has rapidly spread on a global scale. Therefore, it is urgent to identify risk factors that could be associated with severe type of COVID-19 from common type.For this retrospective study, we recruited patients with COVID-19 in Wuhan and Zhoukou. Patients were classified into a severe group and common group based on guidelines after admission. Clinical manifestations and laboratory tests were compared, and univariate binary logistic regression and multivariate regression analyses were applied to assess potential risk factors.A total of 126 patients were recruited from January 23 to March 23, 2020. Ninety cases were identified as the common type and 36 as the severe type. The average age in the severe group was significantly older than that in the common group (P = .008). Patients with severe COVID-19 exhibited higher proportions of dyspnea (P = .001), weakness (P = .023), and diarrhea (P = .046). Moreover, there were more patients with hypertension (P = .01) or coinfection (P = .001) in the severe group than in the common group. Additionally, severe COVID-19 was associated with increased neutrophil counts (P < .001), C-reactive protein (P < .001), procalcitonin (P = .024) and decreased lymphocyte counts (P = .001), hemoglobin (P < .001), total protein (TP) (P < .001), and albumin (ALB) (P < .001). Based on logistic regression analysis, dyspnea (P < .001), TP (P = .042), and ALB (P = .003) were independent risk factors for severe disease.Patients with lower TP, ALB, and dyspnea should be carefully monitored, and early intervention should be implemented to prevent the development of severe disease.


Subject(s)
Coronavirus Infections/diagnosis , Disease Progression , Hospitalization , Pneumonia, Viral/diagnosis , Adult , Aged , Aged, 80 and over , Betacoronavirus , Blood Proteins/analysis , C-Reactive Protein/analysis , COVID-19 , China , Dyspnea/virology , Female , Humans , Male , Middle Aged , Pandemics , Retrospective Studies , Risk Factors , SARS-CoV-2 , Serum Albumin, Human/analysis , Young Adult
20.
Anaesth Crit Care Pain Med ; 39(3): 351-353, 2020 06.
Article in English | MEDLINE | ID: covidwho-986871
SELECTION OF CITATIONS
SEARCH DETAIL