Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 247
Filter
1.
Annals of the Rheumatic Diseases ; 82(Suppl 1):1792, 2023.
Article in English | ProQuest Central | ID: covidwho-20241252

ABSTRACT

BackgroundTofacitinib a small molecule JAK- inhibitors has been approved for use in psoriatic arthritis (PSA) since 2017 while it has shown to be effective in the clinical trials real life data is sparse.With increase in use there has been growing concern about the safety profiles and adverse events which makes it all the more important to have real life data.ObjectivesTo review patient records who were treated with tofacitinib for psoriatic arthritis and to assess the tolerance and continuation rate and also assess the occurrence of adverse events like infections, coronary artery disease.MethodsAll PSA patients who were prescribed tofacitinib from JAN-2021 to JUNE 2022 with minimum of 6 months followup were included for analysis. Demographics, weight recordings, lab parameters and occurence of adverse events were noted.ResultsThere were a total of 71 patients who were prescribed tofacitinib out of which 46 are continuing and 25 have stopped during this period. The mean age was 47.25 (10.9)yrs the mean disease duration was 4.182 (4.474)yrs The reason for stopping tofacitinib was better(52%) followed inefficacy(24%), and miscellaneous(24%)reasons..When analysing before and after tofacitninb one thing whihc was striking is the significant weight gain among patients with minimum of 3.52(3.06) kg weight gain and this weight gain was consistent even in stopped patients.in comparing the lab parameters before and after tofacitininb there was a significant redcution in CRP,ESR,PLATELET COUNT Table 1 and a minimal but insginificant rise in liver enzymes within the physiological range.When compared to before and after tofacitinib there was increased occurence of fatigue(18.3%), minor infections(11.2%), Gastrointerstinal adverse events (11.2%), alopecia (11.2%), Itching(10.4%), headache(9.8%), UTI(5.6%), cough (4.2%), transaminitis(2.8%), covid(1.7%), zoster(1.4%) and CAD(1.4%).ConclusionTofacitinib in psoriatic arthritis is well tolerated with significant reduction in the inflammatory markers and weight gain but serious adverse events in lesser percentage eventhough it leads to significant weight gain.Table 1.PARAMTERSBeforeAfterP valueWeight70.15 (14.19)72.31 (14.24)0.000249ESR45.29 (28.26)35.23 (28.33)0.037CRP21.56 (16.38)10.72 (11.98)<.0001PLATELET COUNT332.92 (88.77)307.09 (88.18)0.0046SGOT30.33 (9.99)35.69 (19.92)0.125SGPT22.57 (12.96)27.98 (20.17)0.116Reference[1]Ly K, Beck KM, Smith MP, Orbai A-M, Liao W. Tofacitinib in the management of active psoriatic arthritis: patient selection and perspectives. Psoriasis (Auckl) [Internet]. 2019;9:97–107. Available from: https://doi.org/10.2147/PTT.S161453Acknowledgements:NIL.Disclosure of InterestsNone Declared.

2.
American Journal of Clinical Pathology, suppl 1 ; 158, 2022.
Article in English | ProQuest Central | ID: covidwho-20232950

ABSTRACT

Introduction To prevent and treat thrombotic complications in patients hospitalized with severe COVID-19 infection, anticoagulation treatments primarily with heparin and low molecular weight heparin have been recommended. Heparin-induced thrombocytopenia (HIT) is a rare but conceivably fatal reaction to heparin that is characterized by a sudden drop in platelet count accompanied by new onset of thrombosis 4-10 days after heparin exposure. The purpose of this retrospective study was to investigate the prevalence of thrombocytopenia and HIT in hospitalized COVID-19 patients, as well as their association with mortality. Methods 3,672 plasma samples were collected from patients admitted to the first wave of COVID-19 in our institution at New York City (March to May 2020). All patients admitted with a platelet count of less than 150 k/ul were assigned to the thrombocytopenic group. In addition, two groups with similar demographics and normal platelet counts were randomly selected based on discharge outcome: alive vs. deceased (n= 88 per group). PF4 IgG Elisa and heparin neutralization were carried out in accordance with the manufacturer's instructions. A positive HIT result required an optical density (OD) greater than 0.4 and heparin neutralization greater than 50%. Statistical analysis was done in R studio (V.1.4.1717) to analyze demographics (age, gender, ethnicity), initial laboratory data, anticoagulation on admission, and thrombosis. Results Only 86 of the 3,672 (2.3%) patients admitted had thrombocytopenia. Only 1 of the 86 patients tested positive for HIT (1.1%). 4 cases of the non-survivors (4.5%) tested positive for HIT compared to none of the survivors in the two groups with normal platelet counts. One of these 4 cases had a history of thrombosis (DVT). Interestingly, the PF4 Elisa ODs in non-survivors were significantly higher than in survivors (0.09 vs. 0.06, p-value< 0.001). Although the platelet count did not differ significantly between the two groups, the mean platelet volume (MPV) on admission and its maximum peak during hospitalization were significantly higher in non-survivors than in survivors. Conclusions We only found HIT positive cases among non-survivors, implying that HIT is associated with COVID severity. The incidence of HIT in severe COVID-19 patients appears to be higher than the pre-COVID-19 historical rates of HIT in hospitalized patients (<1%). Although thrombocytopenia is relatively uncommon in COVID-19 patients, the MPV was significantly higher in non-survivors, suggesting that platelet activation and destruction may explain the higher rate of HIT in COVID-19.

3.
Med. lab ; 26(3): 219-236, 2022. Tabs, ilus, Grafs
Article in Spanish | WHO COVID, LILACS (Americas) | ID: covidwho-20244331

ABSTRACT

El virus SARS-CoV-2 continúa infectando a millones de individuos en el mundo. Aunque los síntomas más frecuentes observados en los pacientes con COVID-19 son fiebre, fatiga y tos, en los casos severos la hipercoagulabilidad y la inflamación son dos condiciones que pueden producir complicaciones y causar daño en órganos, poniendo en riesgo la vida del paciente. Con el fin de clasificar a los pacientes durante el triaje, se han explorado diferentes marcadores hematológicos, incluidos el recuento de plaquetas, linfocitos y eosinófilos, y la relación neutrófilos/ linfocitos, entre otros. Por su parte, para la evaluación de las coagulopatías, se vienen determinando marcadores como el dímero D y el fibrinógeno. En esta revisión se abordan las coagulopatías y los parámetros hematológicos en pacientes con COVID-19, al igual que las anormalidades en la coagulación como la trombocitopenia trombótica inmune inducida por las vacunas contra el SARS-CoV-2


The SARS-CoV-2 virus continues to infect millions of individuals around the world. Although the most frequent symptoms observed in patients with COVID-19 are fever, fatigue and cough, in severe cases hypercoagulability and inflammation are two conditions that can cause complications and organ failure, putting the patient's life at risk. In order to classify patients during triage, different hematological markers have been explored, including platelet, lymphocyte, and eosinophil counts, and the neutrophil/lymphocyte ratio, among others. Furthermore, for the evaluation of coagulopathies, markers such as D-dimer and fibrinogen are being evaluated. This review addresses the coagulopathies and hematological parameters in patients with COVID-19, as well as coagulation abnormalities such as immune thrombotic thrombocytopenia induced by SARS-CoV-2 vaccines


Subject(s)
Humans , COVID-19 , Prognosis , Reference Standards , Thrombosis , Blood Coagulation , Blood Coagulation Disorders , Blood Platelets , Vaccines , Antigens, Differentiation , SARS-CoV-2 , Hematology
4.
J Thromb Haemost ; 2023 May 16.
Article in English | MEDLINE | ID: covidwho-20237345

ABSTRACT

BACKGROUND: Respiratory failure is the primary cause of death in patients with COVID-19, whereas coagulopathy is associated with excessive inflammation and multiorgan failure. Neutrophil extracellular traps (NETs) may exacerbate inflammation and provide a scaffold for thrombus formation. OBJECTIVES: The goal of this study was to determine whether degradation of NETs by recombinant human DNase-I (rhDNase), a safe, Food and Drug Administration-approved drug, reduces excessive inflammation, reverses aberrant coagulation, and improves pulmonary perfusion after experimental acute respiratory distress syndrome (ARDS). METHODS: Intranasal poly(I:C), a synthetic double-stranded RNA, was administered to adult mice for 3 consecutive days to simulate a viral infection, and these subjects were randomized to treatment arms, which received either an intravenous placebo or rhDNase. The effects of rhDNase on immune activation, platelet aggregation, and coagulation were assessed in mice and donor human blood. RESULTS: NETs were observed in bronchoalveolar lavage fluid and within regions of hypoxic lung tissue after experimental ARDS. The administration of rhDNase mitigated peribronchiolar, perivascular, and interstitial inflammation induced by poly(I:C). In parallel, rhDNase degraded NETs, attenuated platelet-NET aggregates, reduced platelet activation, and normalized the clotting time to improve regional perfusion, as observed using gross morphology, histology, and microcomputed tomographic imaging in mice. Similarly, rhDNase reduced NETs and attenuated platelet activation in human blood. CONCLUSION: NETs exacerbate inflammation and promote aberrant coagulation by providing a scaffold for aggregated platelets after experimental ARDS. Intravenous administration of rhDNase degrades NETs and attenuates coagulopathy in ARDS, providing a promising translational approach to improve pulmonary structure and function after ARDS.

5.
Front Immunol ; 14: 1188079, 2023.
Article in English | MEDLINE | ID: covidwho-20237314

ABSTRACT

Background: Immune cell recruitment, endothelial cell barrier disruption, and platelet activation are hallmarks of lung injuries caused by COVID-19 or other insults which can result in acute respiratory distress syndrome (ARDS). Basement membrane (BM) disruption is commonly observed in ARDS, however, the role of newly generated bioactive BM fragments is mostly unknown. Here, we investigate the role of endostatin, a fragment of the BM protein collagen XVIIIα1, on ARDS associated cellular functions such as neutrophil recruitment, endothelial cell barrier integrity, and platelet aggregation in vitro. Methods: In our study we analyzed endostatin in plasma and post-mortem lung specimens of patients with COVID-19 and non-COVID-19 ARDS. Functionally, we investigated the effect of endostatin on neutrophil activation and migration, platelet aggregation, and endothelial barrier function in vitro. Additionally, we performed correlation analysis for endostatin and other critical plasma parameters. Results: We observed increased plasma levels of endostatin in our COVID-19 and non-COVID-19 ARDS cohort. Immunohistochemical staining of ARDS lung sections depicted BM disruption, alongside immunoreactivity for endostatin in proximity to immune cells, endothelial cells, and fibrinous clots. Functionally, endostatin enhanced the activity of neutrophils, and platelets, and the thrombin-induced microvascular barrier disruption. Finally, we showed a positive correlation of endostatin with soluble disease markers VE-Cadherin, c-reactive protein (CRP), fibrinogen, and interleukin (IL)-6 in our COVID-19 cohort. Conclusion: The cumulative effects of endostatin on propagating neutrophil chemotaxis, platelet aggregation, and endothelial cell barrier disruption may suggest endostatin as a link between those cellular events in ARDS pathology.


Subject(s)
COVID-19 , Respiratory Distress Syndrome , Humans , Endostatins/adverse effects , Endostatins/metabolism , Capillary Permeability , Endothelial Cells/metabolism , COVID-19/metabolism , Respiratory Distress Syndrome/pathology , Inflammation/metabolism
6.
Viruses ; 15(5)2023 04 28.
Article in English | MEDLINE | ID: covidwho-20236415

ABSTRACT

BACKGROUND: Mitochondrial dysfunction and redox cellular imbalance indicate crucial function in COVID-19 pathogenesis. Since 11 March 2020, a global pandemic, health crisis and economic disruption has been caused by SARS-CoV-2 virus. Vaccination is considered one of the most effective strategies for preventing viral infection. We tested the hypothesis that preventive vaccination affects the reduced bioenergetics of platelet mitochondria and the biosynthesis of endogenous coenzyme Q10 (CoQ10) in patients with post-acute COVID-19. MATERIAL AND METHODS: 10 vaccinated patients with post-acute COVID-19 (V + PAC19) and 10 unvaccinated patients with post-acute COVID-19 (PAC19) were included in the study. The control group (C) consisted of 16 healthy volunteers. Platelet mitochondrial bioenergy function was determined with HRR method. CoQ10, γ-tocopherol, α-tocopherol and ß-carotene were determined by HPLC, TBARS (thiobarbituric acid reactive substances) were determined spectrophotometrically. RESULTS: Vaccination protected platelet mitochondrial bioenergy function but not endogenous CoQ10 levels, in patients with post-acute COVID-19. CONCLUSIONS: Vaccination against SARS-CoV-2 virus infection prevented the reduction of platelet mitochondrial respiration and energy production. The mechanism of suppression of CoQ10 levels by SARS-CoV-2 virus is not fully known. Methods for the determination of CoQ10 and HRR can be used for monitoring of mitochondrial bioenergetics and targeted therapy of patients with post-acute COVID-19.


Subject(s)
COVID-19 , Humans , COVID-19/prevention & control , SARS-CoV-2 , Oxidation-Reduction , Mitochondria , Vaccination
7.
Revista Medica del Instituto Mexicano del Seguro Social ; 61(3):342-347, 2023.
Article in Spanish | MEDLINE | ID: covidwho-2322124

ABSTRACT

Background: Acute kidney injury (AKI) is frequent in sepsis (25 to 51%), with high mortality (40 to 80%) and long-term complications. Despite its importance we do not have accessible markers in intensive care. In other entities (post-surgical and COVID-19) the neutrophil/lymphocyte and platelet (N/LP) ratio has been associated with acute kidney injury;however, this relationship has not been studied in a pathology with a severe inflammatory response such as sepsis. Objective: To demonstrate the association between N/LP with AKI secondary to sepsis in intensive care. Material and methods: Ambispective cohort study in patients over 18 years who were admitted to intensive care with a diagnosis of sepsis. The N/LP ratio was calculated from admission up to the seventh day and up to the diagnosis of AKI and outcome. Statistical analysis was performed with chi squared test, Cramer's V and multivariate logistic regression. Results: Out of the 239 patients studied, the incidence of AKI developed in 70%. 80.9% of patients with N/LP ratio > 3 had AKI (p < 0.0001, Cramer's V 0.458, OR 3.05, 95% CI 1.602-5.8) and increased renal replacement therapy (21.1 vs. 11.1%, p = 0.043). Conclusion: N/LP ratio > 3 has a moderate association with AKI secondary to sepsis in the intensive care unit. Copyright © 2023 Revista Medica del Instituto Mexicano del Seguro Social.

8.
Saglik Bilimlerinde Ileri Arastirmalar Dergisi / Journal of Advanced Research in Health Sciences ; 5(3):179-185, 2022.
Article in Turkish | CAB Abstracts | ID: covidwho-2321492

ABSTRACT

Objective: The Covid-19 pandemic has revealed the importance of an evidence-based efficient triage system in the early identification of high risk patients and the rational use of limited medical resources for reducing mortality. The aim of this study was to evaluate the role of various inflammatory indices that can be easily calculated using readily accessible, inexpensive routine test parameters in risk stratification and prediction of prognosis in patients with Covid-19. Material and Methods: The study was carried out retrospectively with the data of 8036 patients with Covid-19, who were grouped according to their prognosis in outpatient and inpatient follow-ups, and inpatients as survivors and death. Using the complete blood count and C-reactive protein baseline results of the patients at admission, neutrophillymphocyte ratio (NLR), platelet-lymphocyte ratio (PLR), monocytelymphocyte ratio (MLR), MVP-platelet ratio (MPR), platelet mass index (PMI), systemic immune-inflammatory index (SII), systemic inflammatory response index (SIRI), and multi-inflammatory indices (MII) were calculated. Results: Our results demonstrate that almost all of the inflammatory indices were significantly different in severe patients and in patients with high mortality risk, but not all of them had a predictive value. It has been seen that the most effective factors in determining the disease severity at the onset of Covid-19 are SIRI and age, and SII, MII-1 and MII-3 may also contribute to this prediction. Our results have also revealed that NLR is the most effective independent factor to predict mortality both at disease onset and for inpatients. Conclusion: Inflammatory indices, especially SIRI, NLR, SII, MII-1 and MII-3 can substantially contribute to clinical decisions in the early identification of high-risk patients and predicting mortality beginning from the onset of Covid-19.

9.
Saglik Bilimleri Tip Dergisi, Firat Universitesi ; 36(2):117-124, 2022.
Article in English | GIM | ID: covidwho-2317848

ABSTRACT

Objective: To represent the effects of the severity of COVID-19 infection on platelet large cell ratio (PLC-R). Materials and Methods: A hundred eleven patients diagnosed with COVID-19 were included in this study. Positive results for SARS-CoV-2 based on a typical RT-PCR test performed on nasopharyngeal swabs were included in the study Groups. Patients with COVID-19 were divided into three Groups according to their chest CT features. Group 1 (45 patients) was defined as mild, Group 2 (34 patients) as moderate and Group 3 (32 patients) as severe. Complete blood count parameters including platelet volume indices (PVI) values, CRP, D-dimer and lipid profiles were analyzed in all study participants. The correlation between COVID-19 patient Groups and PLC-R values were demonstrated using SPSS and ANFC methods. Results: The significant impact of our study is that PLC-R was significantly higher in the severe COVID-19 patients than the moderate and mild patients. Spearman's rho correlation analysis showed that PLC-R and WBC levels increased, and Htc and Hb levels decreased with the severity of the disease. ROC analysis showed that PLC-R > 38.3% had 59.4% sensitivity and 68.4% specificity in predicting severe COVID-19 disease (AUC 0.672, %95 CI 0.560, 0.784;p=0.005, cut off=38.3). CRP, ferritin and D-dimer values of the patients in Group 3 were significantly higher than the patients in Group 1, and the iron values of the patients in Group 3 were significantly lower than the patients in Group 1. Conclusion: PLC-R values are useful for anticipating acute thrombotic events. Based on the results of our study, PLC-R values can be used as appropriate biomarkers to describe the severity of COVID-19 infection.

10.
Pakistan Journal of Medical Sciences Quarterly ; 39(3):795, 2023.
Article in English | ProQuest Central | ID: covidwho-2317565

ABSTRACT

Objective: To evaluate the efficacy of hematological parameters to predict severity of COVID-19 patients. Method: This was a cross-sectional comparative study conducted at Central Park Teaching Hospital, Lahore in COVID ward and COVID ICU between April 23, 2021 to June 23, 2021. Patients of all ages and both genders with positive PCR admitted in the COVID ward and ICU during this time span of two months were included in the study. Data was collected retrospectively. Results: This study included 50 patients with male to female ratio of 1.38:1. Though males are more affected by COVID-19 but the difference is not statistically significant. The mean age of the study population was 56.21 and the patients in the severe disease group have higher age. It was observed that in severe/critical group the mean values of total leukocyte count 21.76×103 µI (p-value= 0.002), absolute neutrophil count 71.37% (p-value=0.045), neutrophil lymphocyte ratio (NLR) 12.80 (p-value=0.00) and PT 11.9 seconds (p-value=0.034) and the difference was statistically significant. While in severe/critical group, the mean values of hemoglobin 12.03g/dl (p-value=0.075), lymphocyte count 28.41% (p-value=0.8), platelet count 226×103 µI (p-value=0.67) and APTT 30.7 (p-value=0.081) and the difference was not significantly different between groups. Conclusion: It can be concluded from the study that total leucocyte count, absolute neutrophil count and neutrophil lymphocyte ratio can predict in-hospital mortality and morbidity in COVID-19 patients.

12.
Circ Res ; 132(10): 1272-1289, 2023 05 12.
Article in English | MEDLINE | ID: covidwho-2319061

ABSTRACT

COVID-19 is characterized by dysregulated thrombosis and coagulation that can increase mortality in patients. Platelets are fast responders to pathogen presence, alerting the surrounding immune cells and contributing to thrombosis and intravascular coagulation. The SARS-CoV-2 genome has been found in platelets from patients with COVID-19, and its coverage varies according to the method of detection, suggesting direct interaction of the virus with these cells. Antibodies against Spike and Nucleocapsid have confirmed this platelet-viral interaction. This review discusses the immune, prothrombotic, and procoagulant characteristics of platelets observed in patients with COVID-19. We outline the direct and indirect interaction of platelets with SARS-CoV-2, the contribution of the virus to programmed cell death pathway activation in platelets and the consequent extracellular vesicle release. We discuss platelet activation and immunothrombosis in patients with COVID-19, the effect of Spike on platelets, and possible activation of platelets by classical platelet activation triggers as well as contribution of platelets to complement activation. As COVID-19-mediated thrombosis and coagulation are still not well understood in vivo, we discuss available murine models and mouse adaptable strains.


Subject(s)
COVID-19 , Thrombosis , Mice , Animals , COVID-19/metabolism , SARS-CoV-2 , Blood Platelets/metabolism , Platelet Activation
13.
J Thromb Haemost ; 2023 May 11.
Article in English | MEDLINE | ID: covidwho-2317687

ABSTRACT

Long COVID is a public health emergency affecting millions of people worldwide, characterized by heterogeneous symptoms across multiple organ systems. Here, we discuss the current evidence linking thromboinflammation to postacute sequelae of COVID-19. Studies have found persistence of vascular damage with increased circulating markers of endothelial dysfunction, coagulation abnormalities with heightened thrombin generation capacity, and abnormalities in platelet counts in postacute sequelae of COVID-19. Neutrophil phenotype resembles acute COVID-19 with an increase in activation and Neutrophil Extracellular Trap formation. These insights are potentially linked by elevated platelet-neutrophil aggregate formation. This hypercoagulable state in turn can lead to microvascular thrombosis, evidenced by microclots and elevated D-dimer in the circulation as well as perfusion abnormalities in the lungs and brains of patients with long COVID. Also, COVID-19 survivors experience an increased rate of arterial and venous thrombotic events. We discuss 3 important, potentially intertwined hypotheses that might contribute to thromboinflammation in long COVID: lasting structural changes, most prominently endothelial damage, caused during initial infection; a persistent viral reservoir; and immunopathology driven by a misguided immune system. Finally, we outline the necessity for large, well-characterized clinical cohorts and mechanistic studies to clarify the contribution of thromboinflammation to long COVID.

15.
Ann Hematol ; 102(6): 1307-1322, 2023 Jun.
Article in English | MEDLINE | ID: covidwho-2303196

ABSTRACT

The coagulation, fibrinolytic, anticoagulation, and complement systems are in delicate balance with the vessel wall endothelium ensuring appropriate hemostasis. Coagulopathy in coronavirus disease 2019 (COVID-19) is not a simple disorder of one hemostatic component but a complicated process affecting most of the hemostasis system. COVID-19 disturbs the balance between the procoagulant systems and the regulatory mechanisms. Here, we investigate the effect of COVID-19 on key hemostatic components, including platelets, endothelial cells, coagulation factors, fibrinolytic system, anticoagulant protein system, and complement system, to improve our understanding of the pathophysiological processes underlying COVID-19 coagulopathy based on evidence.


Subject(s)
Blood Coagulation Disorders , COVID-19 , Hemostatics , Humans , Hemostatics/pharmacology , Endothelial Cells/metabolism , Hemostasis , Blood Coagulation Factors/metabolism , Blood Platelets/metabolism , Endothelium, Vascular/metabolism , Fibrinolysis
16.
Biomolecules ; 13(4)2023 04 20.
Article in English | MEDLINE | ID: covidwho-2291408

ABSTRACT

Atherosclerosis is a progressive vascular multifactorial process. The mechanisms underlining the initiating event of atheromatous plaque formation are inflammation and oxidation. Among the modifiable risk factors for cardiovascular diseases, diet and especially the Mediterranean diet (MedDiet), has been widely recognized as one of the healthiest dietary patterns. Olive oil (OO), the main source of the fatty components of the MedDiet is superior to the other "Mono-unsaturated fatty acids containing oils" due to the existence of specific microconstituents. In this review, the effects of OO microconstituents in atherosclerosis, based on data from in vitro and in vivo studies with special attention on their inhibitory activity against PAF (Platelet-Activating Factor) actions, are presented and critically discussed. In conclusion, we propose that the anti-atherogenic effect of OO is attributed to the synergistic action of its microconstituents, mainly polar lipids that act as PAF inhibitors, specific polyphenols and α-tocopherol that also exert anti-PAF activity. This beneficial effect, also mediated through anti-PAF action, can occur from microconstituents extracted from olive pomace, a toxic by-product of the OO production process that constitutes a significant ecological problem. Daily intake of moderate amounts of OO consumed in the context of a balanced diet is significant for healthy adults.


Subject(s)
Atherosclerosis , Olea , Adult , Humans , Olive Oil/pharmacology , Plant Oils/pharmacology , Risk Factors , Atherosclerosis/drug therapy , Atherosclerosis/prevention & control
17.
Microorganisms ; 11(4)2023 Apr 06.
Article in English | MEDLINE | ID: covidwho-2290795

ABSTRACT

People living with HIV (PLWHIV) present an increased risk of adverse cardiovascular events. We aimed to assess whether antiretroviral therapy (ART) pharmacologically enhances platelet reactivity and platelet activation intensity, and explore the potential association with underlying inflammatory status. This was a cross-sectional cohort study carried out among PLWHIV on diverse ART regimens. Platelet reactivity and activation intensity were assessed using the bedside point-of-care VerifyNow assay, in P2Y12 reaction units (PRU), measurements of monocyte-platelet complexes, and P-selectin and GPIIb/IIIa expression increase, following activation with ADP, respectively. Levels of major inflammatory markers and whole blood parameters were also evaluated. In total, 71 PLWHIV, 59 on ART and 22 healthy controls, were included in this study. PRU values were significantly elevated in PLWHIV compared to controls [Mean; 257.85 vs. 196.67, p < 0.0001], but no significant differences were noted between ART-naïve or ART-experienced PLWHIV, or between TAF/TDF and ABC based regimens, similar to systemic inflammatory response. However, within-group analysis showed that PRUs were significantly higher in ABC/PI vs ABC/INSTI or TAF/TDF + PI patients, in line with levels of IL-2. PRU values did not correlate strongly with CD4 counts, viral load, or cytokine values. P-selectin and GPIIb/IIIa expression increased following ADP activation and were significantly more prominent in PLWHIV (p < 0.005). Platelet reactivity and platelet activation intensity were shown to be increased in PLWHIV, but they did not appear to be related to ART initiation, similar to the underlying systemic inflammatory response.

18.
Biomedicines ; 11(4)2023 Apr 15.
Article in English | MEDLINE | ID: covidwho-2294897

ABSTRACT

Although there is increasing evidence that oxidative stress and inflammation induced by COVID-19 may contribute to increased risk and severity of thromboses, the underlying mechanism(s) remain to be understood. The purpose of this review is to highlight the role of blood lipids in association with thrombosis events observed in COVID-19 patients. Among different types of phospholipases A2 that target cell membrane phospholipids, there is increasing focus on the inflammatory secretory phospholipase A2 IIA (sPLA2-IIA), which is associated with the severity of COVID-19. Analysis indicates increased sPLA2-IIA levels together with eicosanoids in the sera of COVID patients. sPLA2 could metabolise phospholipids in platelets, erythrocytes, and endothelial cells to produce arachidonic acid (ARA) and lysophospholipids. Arachidonic acid in platelets is metabolised to prostaglandin H2 and thromboxane A2, known for their pro-coagulation and vasoconstrictive properties. Lysophospholipids, such as lysophosphatidylcholine, could be metabolised by autotaxin (ATX) and further converted to lysophosphatidic acid (LPA). Increased ATX has been found in the serum of patients with COVID-19, and LPA has recently been found to induce NETosis, a clotting mechanism triggered by the release of extracellular fibres from neutrophils and a key feature of the COVID-19 hypercoagulable state. PLA2 could also catalyse the formation of platelet activating factor (PAF) from membrane ether phospholipids. Many of the above lipid mediators are increased in the blood of patients with COVID-19. Together, findings from analyses of blood lipids in COVID-19 patients suggest an important role for metabolites of sPLA2-IIA in COVID-19-associated coagulopathy (CAC).

19.
Front Immunol ; 14: 1151926, 2023.
Article in English | MEDLINE | ID: covidwho-2306444

ABSTRACT

Thrombosis is a frequent cause of cardiovascular mortality and hospitalization. Current antithrombotic strategies, however, target both thrombosis and physiological hemostasis and thereby increase bleeding risk. In recent years the pathophysiological understanding of thrombus formation has significantly advanced and inflammation has become a crucial element. Neutrophils as most frequent immune cells in the blood and their released mediators play a key role herein. Neutrophil-derived cathelicidin next to its strong antimicrobial properties has also shown to modulates thrombosis and thus presents a potential therapeutic target. In this article we review direct and indirect (immune- and endothelial cell-mediated) effects of cathelicidin on platelets and the coagulation system. Further we discuss its implications for large vessel thrombosis and consecutive thromboinflammation as well as immunothrombosis in sepsis and COVID-19 and give an outlook for potential therapeutic prospects.


Subject(s)
COVID-19 , Thrombosis , Humans , Thrombosis/drug therapy , Thrombosis/etiology , Thromboinflammation , Inflammation/drug therapy , Cathelicidins
20.
Journal of Cardiovascular Disease Research ; 13(8):2321-2329, 2022.
Article in English | GIM | ID: covidwho-2266121

ABSTRACT

Background: In India, the first case of COVID-19 was reported on January 30, 2020. The case reporting is based on the testing of individuals by Real-time Reverse Transcription- Polymerase Chain Reaction (RT-qPCR). The present study was conducted to evaluatedifferent parameters, Haematological and Biomarker variations in patients with SARS-CoV2 Infection to assess the prognostic significance. Material & Methods: The present prospective study was conducted among 70 patients who were diagnosed with COVID-19 infection. Relavant physical examination and clinical data of the patient and routine blood investigations including, CBC, serum biochemistry, coagulation function and measurement of inflammatory markers were performed. The results were analyzed by using a SPSS Statistics software version 25.0. Results: In the present study total patients were 70 out of which 58.6% were males and 41.4% were females. Maximum subjects belong to age group 61-80 yrs (47.1%). Mean haemoglobin was 12.89g/l, mean platelet was 9.96x103/l. Mean neutrophil were 88.21%, mean lymphocyte were 8.84%, mean eosinophil were 1.47%, mean monocyte was 1.59%, mean TLC was 12007.14/l. Mean random blood sugar was 148.09 mg/dl. Mean D-dimer was 0.56. Mean CRP levels were 65.5 mg/l. Mean LDH was 516.03 IU/L, mean IL-6 was 282.6pg/ml, and mean procalcitonin was 0.8 ng/ml. Mean SGOT was 62.36u/l, mean ALP was 171.87IU/L, mean urea levels were 57.10 mg/dl and mean INR was 1.22. Outcome mortality was present in total 14 subjects (5 were male and 9 were female) out of all 70 subjects. Conclusion: The present study concluded that Mean values of neutrophil, eosinophil, TLC, random blood sugar, IL6, SGOT, ALP, urea levels and INR were increased in patients with SARS-CoV2 Infection.

SELECTION OF CITATIONS
SEARCH DETAIL