Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.551
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 21(11): 1319-1326, 2020 11.
Article in English | MEDLINE | ID: mdl-33077953

ABSTRACT

Injury is a key driver of inflammation, a critical yet necessary response involving several mediators that is aimed at restoring tissue homeostasis. Inflammation in the central nervous system can be triggered by a variety of stimuli, some intrinsic to the brain and others arising from peripheral signals. Fine-tuned regulation of this response is crucial in a system that is vulnerable due to, for example, aging and ongoing neurodegeneration. In this context, seemingly harmless interventions like a common surgery to repair a broken limb can overwhelm the immune system and become the driver of further complications such as delirium and other perioperative neurocognitive disorders. Here, we discuss potential mechanisms by which the immune system affects the central nervous system after surgical trauma. Together, these neuroimmune interactions are becoming hallmarks of and potential therapeutic targets for multiple neurologic conditions, including those affecting the perioperative space.


Subject(s)
Inflammation/etiology , Inflammation/metabolism , Nervous System Diseases/etiology , Nervous System Diseases/metabolism , Postoperative Complications , Alarmins/genetics , Alarmins/metabolism , Animals , Blood Coagulation , Blood Coagulation Factors/genetics , Blood Coagulation Factors/metabolism , Blood-Brain Barrier/metabolism , Combined Modality Therapy , Complement Activation/immunology , Complement System Proteins/immunology , Complement System Proteins/metabolism , Disease Management , Disease Susceptibility , Humans , Immune System/immunology , Immune System/metabolism , Inflammation/diagnosis , Inflammation/therapy , Nervous System Diseases/diagnosis , Nervous System Diseases/therapy , Neuroglia/immunology , Neuroglia/metabolism , Neuroimmunomodulation , Treatment Outcome
2.
Brain Behav Immun ; 119: 188-196, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38555993

ABSTRACT

INTRODUCTION: Negative symptoms impact the quality of life of individuals with psychosis and current treatment options for negative symptoms have limited effectiveness. Previous studies have demonstrated that complement and coagulation pathway protein levels are related to later psychotic experiences, psychotic disorder, and functioning. However, the prognostic relationship between complement and coagulation proteins and negative symptoms is poorly characterised. METHODS: In the North American Prodrome Longitudinal Studies 2 and 3, negative symptoms in 431 individuals at clinical high-risk for psychosis (mean age: 18.2, SD 3.6; 42.5 % female) were measured at multiple visits over 2 years using the Scale of Psychosis-Risk Symptoms. Plasma proteins were quantified at baseline using mass spectrometry. Four factors were derived to represent levels of proteins involved in the activation or regulation of the complement or coagulation systems. The relationships between standardised protein group factors and serial measurements of negative symptoms over time were modelled using generalised least squares regression. Analyses were adjusted for baseline candidate prognostic factors: negative symptoms, positive symptoms, functioning, depressive symptoms, suicidal ideation, cannabis use, tobacco use, antipsychotic use, antidepressant use, age, and sex. RESULTS: Clinical and demographic prognostic factors of follow-up negative symptoms included negative, positive, and depressive symptoms, functioning, and age. Adjusting for all candidate prognostic factors, the complement regulators group and the coagulation regulators group were identified as prognostic factors of follow-up negative symptoms (ß: 0.501, 95 % CI: 0.160, 0.842; ß: 0.430, 95 % CI: 0.080, 0.780 respectively. The relationship between complement regulator levels and negative symptoms was also observed in NAPLS2 alone (ß: 0.501, 95 % CI: -0.037, 1.039) and NAPLS3 alone, additionally adjusting for BMI (ß: 0.442, 95 % CI: 0.127, 0.757). CONCLUSION: The results indicate that plasma complement and coagulation regulator levels are prognostic factors of negative symptoms, independent of clinical and demographic prognostic factors. These results suggest complement and coagulation regulator levels could have potential utility in informing treatment decisions for negative symptoms in individuals at risk.


Subject(s)
Complement System Proteins , Psychotic Disorders , Humans , Female , Male , Prognosis , Adolescent , Young Adult , Complement System Proteins/metabolism , Complement System Proteins/analysis , Psychotic Disorders/blood , Adult , Blood Coagulation Factors/metabolism , Blood Coagulation Factors/analysis , Longitudinal Studies
3.
Biochemistry (Mosc) ; 89(Suppl 1): S14-S33, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38621742

ABSTRACT

Reactive oxygen species (ROS) are constantly generated in a living organism. An imbalance between the amount of generated reactive species in the body and their destruction leads to the development of oxidative stress. Proteins are extremely vulnerable targets for ROS molecules, which can cause oxidative modifications of amino acid residues, thus altering structure and function of intra- and extracellular proteins. The current review considers the effect of oxidation on the structural rearrangements and functional activity of hemostasis proteins: coagulation system proteins such as fibrinogen, prothrombin/thrombin, factor VII/VIIa; anticoagulant proteins - thrombomodulin and protein C; proteins of the fibrinolytic system such as plasminogen, tissue plasminogen activator and plasminogen activator inhibitor-1. Structure and function of the proteins, oxidative modifications, and their detrimental consequences resulting from the induced oxidation or oxidative stress in vivo are described. Possible effects of oxidative modifications of proteins in vitro and in vivo leading to disruption of the coagulation and fibrinolysis processes are summarized and systematized, and the possibility of a compensatory mechanism in maintaining hemostasis under oxidative stress is analyzed.


Subject(s)
Hemostasis , Tissue Plasminogen Activator , Tissue Plasminogen Activator/metabolism , Reactive Oxygen Species , Blood Coagulation , Blood Coagulation Factors/metabolism , Oxidative Stress
5.
Ann Hematol ; 102(7): 1939-1949, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37226021

ABSTRACT

Yellow fever (YF) is an acute tropical infectious disease caused by an arbovirus and can manifest as a classic hemorrhagic fever. The mechanism of the bleeding diathesis in YF is not well understood. We assessed clinical and laboratory data (including a panel of coagulation tests) from 46 patients with moderate (M) and severe (S) YF admitted to a local hospital between January 2018 and April 2018. Among 46 patients, 34 had SYF of whom 12 (35%) patients died. A total of 21 (45%) patients developed some type of bleeding manifestation and 15 (32%) presented severe bleeding. Patients with SYF had more severe thrombocytopenia (p = 0.001); prolonged activated partial thromboplastin time (aPTT) and thrombin time (TT) (p = 0.03 and p = 0.005, respectively); reduced plasma levels of coagulation factor (F) II (p < 0.01), FIX (p = 0.01), and FX (p = 0.04); and D-dimer levels almost 10 times higher (p < 0.01) when compared with patients with MYF. Patients who died had more bleeding (p = 0.03), more major bleeding (p = 0.03), prolonged international normalized ratio (INR) and aPTT (p = 0.003 and p = 0.002, respectively), as well as lower activity of FII (p = 0.02), FV (p = 0.001), FVII (p = 0.005), FIX (p = 0.01), and protein C (p = 0.01) than the ones who survived. FVIII levels were either normal or increased in all patients studied. Our results suggest that the bleeding diathesis of SYF is associated with the deficiency of coagulation factors produced by the liver. Prolonged INR and aPTT and reduced FII, FV, FVII, FIX, and protein C were associated with death.


Subject(s)
Blood Coagulation Disorders , Yellow Fever , Humans , Protein C , Disease Susceptibility , Blood Coagulation Factors/metabolism , Blood Coagulation Tests/methods
6.
Ann Hematol ; 102(6): 1307-1322, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37074380

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
7.
BMC Gastroenterol ; 23(1): 65, 2023 Mar 09.
Article in English | MEDLINE | ID: mdl-36894870

ABSTRACT

BACKGROUND: Current quantitative approaches to assess chronic liver disease (CLD) severity have limitations. Further, portal vein thrombosis (PVT) pre-liver transplant (LT) is a major contributor to morbidity in CLD; the means of detecting and/or predicting PVT are limited. We sought to explore whether plasma coagulation factor activity levels can serve as a substitute for prothrombin time/international normalized ratio (PT/INR) in the Model for End-stage Liver Disease (MELD), and/or help assess the risk of PVT. METHODS: Plasma activity levels of Factor V (FV), Factor VIII (FVIII), Protein C (PC), and Protein S (PS) and the concentrations of D-dimer, sP-selectin, and asTF were assessed in two cohorts of CLD patients (ambulatory, n = 42; LT, n = 43). RESULTS: FV and PC activity levels strongly correlated with MELD scores, which enabled the development of a novel scoring system based on multiple linear regressions of the correlations of FV and PC activity with MELD-Na that substitutes PT/INR. Six-month and 1-year follow-up revealed that our novel approach was non-inferior to MELD-Na at predicting mortality. A significant inverse correlation between FVIII activity levels and PVT was found in the LT cohort (p = 0.010); FV and PS activity levels were in-trend (p = 0.069, p = 0.064). We developed a logistic regression-based compensation score to identify patients at risk of PVT. CONCLUSIONS: We demonstrate that FV and PC activity levels may be used to replace PT/INR in MELD scoring. We also show the potential of using the combination of FV, FVIII, and PS activity levels to assess the risk of PVT in CLD.


Subject(s)
End Stage Liver Disease , Liver Diseases , Venous Thrombosis , Humans , Portal Vein/pathology , Liver Cirrhosis , End Stage Liver Disease/complications , End Stage Liver Disease/surgery , Severity of Illness Index , Liver Diseases/complications , Liver Diseases/pathology , Blood Coagulation Factors/metabolism , Venous Thrombosis/diagnosis
8.
Curr Opin Organ Transplant ; 28(6): 404-411, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37728052

ABSTRACT

PURPOSE OF REVIEW: Rebalanced hemostasis describes the precarious balance of procoagulant and antithrombotic proteins in patients with severe liver failure. This review is aimed to discuss currently available coagulation monitoring tests and pertinent decision-making process for plasma coagulation factor replacements during liver transplantation (LT). RECENT FINDINGS: Contemporary viscoelastic coagulation monitoring systems have demonstrated advantages over conventional coagulation tests in assessing the patient's coagulation status and tailoring hemostatic interventions. There is increasing interest in the use of prothrombin complex and fibrinogen concentrates, but it remains to be proven if purified factor concentrates are more efficacious and safer than allogeneic hemostatic components. Furthermore, the decision to use antifibrinolytic therapy necessitates careful considerations given the risks of venous thromboembolism in severe liver failure. SUMMARY: Perioperative hemostatic management and thromboprophylaxis for LT patients is likely to be more precise and patient-specific through a better understanding and monitoring of rebalanced coagulation. Further research is needed to refine the application of these tools and develop more standardized protocols for coagulation management in LT.


Subject(s)
Hemostatics , Liver Transplantation , Humans , Anticoagulants/adverse effects , Anticoagulants/pharmacology , Blood Coagulation Factors/adverse effects , Blood Coagulation Factors/metabolism , Blood Coagulation Factors/pharmacology , Decision Making , Hemostasis , Hemostatics/adverse effects , Hemostatics/pharmacology , Liver Failure , Liver Transplantation/adverse effects , Liver Transplantation/methods , Venous Thromboembolism/drug therapy
9.
J Mol Evol ; 90(6): 418-428, 2022 12.
Article in English | MEDLINE | ID: mdl-36181519

ABSTRACT

Vertebrate blood coagulation is controlled by a cascade containing more than 20 proteins. The cascade proteins are found in the blood in their zymogen forms and when the cascade is triggered by tissue damage, zymogens are activated and in turn activate their downstream proteins by serine protease activity. In this study, we examined proteomes of 21 chordates, of which 18 are vertebrates, to reveal the modular evolution of the blood coagulation cascade. Additionally, two Arthropoda species were used to compare domain arrangements of the proteins belonging to the hemolymph clotting and the blood coagulation cascades. Within the vertebrate coagulation protein set, almost half of the studied proteins are shared with jawless vertebrates. Domain similarity analyses revealed that there are multiple possible evolutionary trajectories for each coagulation protein. During the evolution of higher vertebrate clades, gene and genome duplications led to the formation of other coagulation cascade proteins.


Subject(s)
Blood Coagulation Factors , Chordata , Animals , Blood Coagulation Factors/genetics , Blood Coagulation Factors/metabolism , Vertebrates/genetics , Blood Coagulation/genetics , Chordata/genetics , Genome
10.
Biochem Biophys Res Commun ; 628: 49-56, 2022 11 05.
Article in English | MEDLINE | ID: mdl-36081278

ABSTRACT

The coagulation factor 9 gene (FIX) point mutation contributes to most hemophilia B cases, providing ideal gene correction models. Here we identified the frequent mutation G20519A (R226Q) in FIX, which resulted in many severe and moderate hemophilia B patients. This study aimed to investigate the effect of HDR and base editing in correcting FIX mutant. We first constructed HEK293 and liver-derived cell lines Huh7 cells stabling carrying mutated FIX containing G20519A (HEK293-FIXmut and Huh7-FIXmut). Then, CRISPR/Cas9-based homology-directed repair (HDR) and base editing were used for the correction of this mutated point. We used Cas9 nickase (nCas9) mediated HDR and the advanced base editor ABE8e to correct G20519A and then measured the concentration and activity of FIX. Furthermore, we used the star-shaped poly(lysine) gene nanocarriers to deliver the ABE8e correction systems into HEK293-FIXmut and Huh7-FIXmut stem cells to correct mutated FIX. As a result, we found that gRNAs directed inefficient HDR in correcting G20519A. The ABE8e corrected the mutation efficiently in both HEK293-FIXmut and Huh7-FIXmut stem cells. In addition, the star-shaped poly(lysine) carriers delivered non-viral vectors into stem cells efficiently. The nanocarriers-delivered ABE8e system corrected mutated FIX in stem cells, and the stem cells secreted active FIX in high concentration. In conclusion, our study provides a potential alternative for correcting mutated FIX in hemophilia B patients.


Subject(s)
Gene Editing , Hemophilia A , Hemophilia B , Aminohydrolases/genetics , Blood Coagulation Factors/genetics , Blood Coagulation Factors/metabolism , CRISPR-Cas Systems/genetics , Deoxyribonuclease I/metabolism , Gene Editing/methods , HEK293 Cells , Hemophilia A/genetics , Hemophilia A/metabolism , Hemophilia B/genetics , Hemophilia B/therapy , Humans , Mutation , Mutation, Missense , Polylysine/chemistry , Stem Cells/metabolism
11.
Lipids Health Dis ; 21(1): 143, 2022 Dec 21.
Article in English | MEDLINE | ID: mdl-36544148

ABSTRACT

BACKGROUND: Studies on the associations between lipid parameters and different hemostatic factors in men and women from the general population are scarce. It was therefore examined whether there are possible relationships between routinely measured serum lipids (total cholesterol, HDL-cholesterol, non-HDL-cholesterol, LDL-cholesterol, and triglycerides) and different hemostatic factors (activated partial thromboplastin time (aPTT), fibrinogen, factor VIII, antithrombin III (AT III), protein C, protein S, and D-dimer). METHODS: The analysis was based on data from the Cooperative Health Research in the Region of Augsburg (KORA)-Fit study, which included 805 participants (378 men, 427 women) with a mean age of 63.1 years. Sex-specific associations between serum lipids and coagulation factors were investigated using multivariable linear regression models. RESULTS: In men, total cholesterol was inversely related to aPTT but positively associated with protein C activity. HDL cholesterol was inversely related to aPTT and fibrinogen. LDL cholesterol, non-HDL cholesterol, and triglycerides showed a positive association with protein C and protein S activity. In women, LDL-cholesterol, total cholesterol, and non-HDL-cholesterol were positively related to AT III concentrations and protein C and S activity. Additionally, non-HDL-cholesterol was positively associated with factor VIII activity. HDL cholesterol was inversely related to fibrinogen. Triglycerides showed a positive relationship with protein C activity. CONCLUSIONS: There seem to be sex differences regarding various associations between blood lipid levels and hemostatic factors. Further studies are needed to address the possible impact of these associations on cardiovascular risk and the underlying mechanisms.


Subject(s)
Blood Coagulation Factors , Lipid Metabolism , Sex Characteristics , Female , Humans , Male , Middle Aged , Cholesterol , Cholesterol, HDL , Cholesterol, LDL , Cross-Sectional Studies , Factor VIII , Fibrinogen/analysis , Lipids/blood , Lipoproteins , Protein C , Risk Factors , Triglycerides , Lipid Metabolism/physiology , Blood Coagulation Factors/metabolism
12.
Int J Clin Pract ; 2022: 3144685, 2022.
Article in English | MEDLINE | ID: mdl-35685509

ABSTRACT

Background: Changes of the coagulation system are promoted by serious infectious or noninfectious diseases, surgical procedures, and exogenous substances, including drugs. This study aimed to assess the effect of methylprednisolone pulses on selected parameters of the coagulation system. Methods: The study group consisted of patients suffering from multiple sclerosis, thyroid orbitopathy, or sudden sensorineural hearing loss. 48 patients and 20 healthy volunteers were examined. The hemostatic parameters: activity of coagulation factors (VIII, IX, and XI), antithrombin activity, protein C and S activity, and concentration of soluble tissue factor were analyzed at baseline and after 3 g and 5 g of methylprednisolone administration. Results: A statistically significant increase was noted in the activity of all the studied plasma coagulation factors, plasma coagulation inhibitors (except protein S activity), and the concentration of soluble tissue factor after methylprednisolone administration. Conclusion: The glucocorticoids administered in the intravenous pulses of methylprednisolone shift the balance toward thromboembolic complications.


Subject(s)
Glucocorticoids , Thromboplastin , Blood Coagulation Factors/metabolism , Blood Coagulation Tests , Humans , Methylprednisolone/therapeutic use
13.
Proc Natl Acad Sci U S A ; 116(16): 8038-8047, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30944221

ABSTRACT

Abdominal aortic aneurysm (AAA) is an inflammatory vascular disease with high mortality and limited treatment options. How blood lipids regulate AAA development is unknown. Here lipidomics and genetic models demonstrate a central role for procoagulant enzymatically oxidized phospholipids (eoxPL) in regulating AAA. Specifically, through activating coagulation, eoxPL either promoted or inhibited AAA depending on tissue localization. Ang II administration to ApoE-/- mice increased intravascular coagulation during AAA development. Lipidomics revealed large numbers of eoxPL formed within mouse and human AAA lesions. Deletion of eoxPL-generating enzymes (Alox12 or Alox15) or administration of the factor Xa inhibitor rivaroxaban significantly reduced AAA. Alox-deficient mice displayed constitutively dysregulated hemostasis, including a consumptive coagulopathy, characterized by compensatory increase in prothrombotic aminophospholipids (aPL) in circulating cell membranes. Intravenously administered procoagulant PL caused clotting factor activation and depletion, induced a bleeding defect, and significantly reduced AAA development. These data suggest that Alox deletion reduces AAA through diverting coagulation away from the vessel wall due to eoxPL deficiency, instead activating clotting factor consumption and depletion in the circulation. In mouse whole blood, ∼44 eoxPL molecular species formed within minutes of clot initiation. These were significantly elevated with ApoE-/- deletion, and many were absent in Alox-/- mice, identifying specific eoxPL that modulate AAA. Correlation networks demonstrated eoxPL belonged to subfamilies defined by oxylipin composition. Thus, procoagulant PL regulate AAA development through complex interactions with clotting factors. Modulation of the delicate balance between bleeding and thrombosis within either the vessel wall or circulation was revealed that can either drive or prevent disease development.


Subject(s)
Aorta, Abdominal/physiopathology , Aortic Aneurysm, Abdominal , Phospholipids , Angiotensins/metabolism , Animals , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/physiopathology , Blood Coagulation Factors/genetics , Blood Coagulation Factors/metabolism , Disease Models, Animal , Female , Lipoxygenase/genetics , Lipoxygenase/metabolism , Male , Mice , Mice, Knockout, ApoE , Phospholipids/genetics , Phospholipids/metabolism
14.
Int J Mol Sci ; 23(3)2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35163105

ABSTRACT

To obtain a more detailed picture of macrophage (MΦ) biology, in the current study, we analyzed the transcriptome of mouse peritoneal MΦs by RNA-seq and PCR-based transcriptomics. The results show that peritoneal MΦs, based on mRNA content, under non-inflammatory conditions produce large amounts of a number of antimicrobial proteins such as lysozyme and several complement components. They were also found to be potent producers of several chemokines, including platelet factor 4 (PF4), Ccl6, Ccl9, Cxcl13, and Ccl24, and to express high levels of both TGF-ß1 and TGF-ß2. The liver is considered to be the main producer of most complement and coagulation components. However, we can now show that MΦs are also important sources of such compounds including C1qA, C1qB, C1qC, properdin, C4a, factor H, ficolin, and coagulation factor FV. In addition, FX, FVII, and complement factor B were expressed by the MΦs, altogether indicating that MΦs are important local players in both the complement and coagulation systems. For comparison, we analyzed human peripheral blood monocytes. We show that the human monocytes shared many characteristics with the mouse peritoneal MΦs but that there were also many major differences. Similar to the mouse peritoneal MΦs, the most highly expressed transcript in the monocytes was lysozyme, and high levels of both properdin and ficolin were observed. However, with regard to connective tissue components, such as fibronectin, lubricin, syndecan 3, and extracellular matrix protein 1, which were highly expressed by the peritoneal MΦs, the monocytes almost totally lacked transcripts. In contrast, monocytes expressed high levels of MHC Class II, whereas the peritoneal MΦs showed very low levels of these antigen-presenting molecules. Altogether, the present study provides a novel view of the phenotype of the major MΦ subpopulation in the mouse peritoneum and the large peritoneal MΦs and places the transcriptome profile of the peritoneal MΦs in a broader context, including a comparison of the peritoneal MΦ transcriptome with that of human peripheral blood monocytes and the liver.


Subject(s)
Blood Coagulation Factors/metabolism , Blood Coagulation , Complement System Proteins/immunology , Liver/immunology , Macrophages, Peritoneal/immunology , Monocytes/immunology , Transcriptome , Animals , Complement System Proteins/metabolism , Female , Liver/metabolism , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred BALB C , Monocytes/metabolism
15.
Int J Mol Sci ; 23(4)2022 Feb 19.
Article in English | MEDLINE | ID: mdl-35216433

ABSTRACT

Platelets, which are small anuclear cell fragments, play important roles in thrombosis and hemostasis, but also actively release factors that can both suppress and induce viral infections. Platelet-released factors include sCD40L, microvesicles (MVs), and alpha granules that have the capacity to exert either pro-inflammatory or anti-inflammatory effects depending on the virus. These factors are prime targets for use in extracellular vesicle (EV)-based therapy due to their ability to reduce viral infections and exert anti-inflammatory effects. While there are some studies regarding platelet microvesicle-based (PMV-based) therapy, there is still much to learn about PMVs before such therapy can be used. This review provides the background necessary to understand the roles of platelet-released factors, how these factors might be useful in PMV-based therapy, and a critical discussion of current knowledge of platelets and their role in viral diseases.


Subject(s)
Blood Coagulation Factors/metabolism , Blood Platelets/metabolism , Extracellular Vesicles/metabolism , Virus Diseases/metabolism , Animals , Cell-Derived Microparticles/metabolism , Humans , Platelet Activation/physiology
16.
Molecules ; 27(11)2022 May 29.
Article in English | MEDLINE | ID: mdl-35684431

ABSTRACT

In blood coagulation, circulating platelets and coagulation factors are crucial for the primary process because thrombi are generated by fibrin clotting with fibrinogen, thrombin, FXIIIa, and platelet activation. Therefore, strategies to reduce the activity of key coagulation factors, or interfere with their functions and delay the activation of platelets can be used as important tools to suppress excessive blood clot formation and platelet hyperactivation. This study examined the antithrombotic activity and hematological toxicity of PA, IVA, and 4-HA isolated from M. tricuspidata (Carr.) Bur in several in vitro experiments and inhibitor assays. We found that PA, IVA, and 4-HA attenuated the formation of fibrin polymers/clots and degraded the blood clots. These compounds inhibited the activities of procoagulant proteases and fibrinoligase, and prolonged the coagulation time. There was a significant reduction in platelet function and ATP or serotonin levels in thrombin-activated platelets. An inhibitor study showed that PA exhibited a mixed inhibition type for thrombin, an uncompetitive inhibition type for FXa, and a non-competitive inhibition type for FXIIIa and IVA, while 4-HA exhibited an uncompetitive inhibition type for thrombin and non-competitive inhibition type for FXa and FXIIIa. These three compounds (up to 50 µg/mL) were not toxic to blood cells.


Subject(s)
Maclura , Thrombosis , Blood Coagulation Factors/metabolism , Blood Platelets/metabolism , Fibrin , Fibrinolytic Agents/pharmacology , Humans , Hydroxybenzoates/metabolism , Thrombin/metabolism , Thrombosis/metabolism
17.
Cardiovasc Diabetol ; 20(1): 172, 2021 08 23.
Article in English | MEDLINE | ID: mdl-34425843

ABSTRACT

BACKGROUND: Severe acute respiratory syndrome coronavirus 2 (SARS-Cov-2)-induced infection, the cause of coronavirus disease 2019 (COVID-19), is characterized by acute clinical pathologies, including various coagulopathies that may be accompanied by hypercoagulation and platelet hyperactivation. Recently, a new COVID-19 phenotype has been noted in patients after they have ostensibly recovered from acute COVID-19 symptoms. This new syndrome is commonly termed Long COVID/Post-Acute Sequelae of COVID-19 (PASC). Here we refer to it as Long COVID/PASC. Lingering symptoms persist for as much as 6 months (or longer) after acute infection, where COVID-19 survivors complain of recurring fatigue or muscle weakness, being out of breath, sleep difficulties, and anxiety or depression. Given that blood clots can block microcapillaries and thereby inhibit oxygen exchange, we here investigate if the lingering symptoms that individuals with Long COVID/PASC manifest might be due to the presence of persistent circulating plasma microclots that are resistant to fibrinolysis. METHODS: We use techniques including proteomics and fluorescence microscopy to study plasma samples from healthy individuals, individuals with Type 2 Diabetes Mellitus (T2DM), with acute COVID-19, and those with Long COVID/PASC symptoms. RESULTS: We show that plasma samples from Long COVID/PASC still contain large anomalous (amyloid) deposits (microclots). We also show that these microclots in both acute COVID-19 and Long COVID/PASC plasma samples are resistant to fibrinolysis (compared to plasma from controls and T2DM), even after trypsinisation. After a second trypsinization, the persistent pellet deposits (microclots) were solubilized. We detected various inflammatory molecules that are substantially increased in both the supernatant and trapped in the solubilized pellet deposits of acute COVID-19 and Long COVID/PASC, versus the equivalent volume of fully digested fluid of the control samples and T2DM. Of particular interest was a substantial increase in α(2)-antiplasmin (α2AP), various fibrinogen chains, as well as Serum Amyloid A (SAA) that were trapped in the solubilized fibrinolytic-resistant pellet deposits. CONCLUSIONS: Clotting pathologies in both acute COVID-19 infection and in Long COVID/PASC might benefit from following a regime of continued anticlotting therapy to support the fibrinolytic system function.


Subject(s)
Antifibrinolytic Agents/metabolism , Blood Coagulation Factors/metabolism , COVID-19/complications , Adult , Disease Progression , Female , Humans , Male , Middle Aged , SARS-CoV-2/pathogenicity , Post-Acute COVID-19 Syndrome
18.
Transfusion ; 61(4): 1035-1040, 2021 04.
Article in English | MEDLINE | ID: mdl-33634868

ABSTRACT

BACKGROUND: Cryoprecipitate (CRYO) is neither produced nor supplied by the Japanese Red Cross Society. A novel CRYO extraction method established in-house by modifying a thaw-siphon technique was demonstrated in this study. STUDY DESIGN AND METHODS: A pack of fresh frozen plasma was thawed and equally divided into two bags for CRYO extraction by different methods. CRYO was extracted from the blood plasma using a standard centrifugation method and our modified thaw-siphon method (Bokutoh-siphon method; B method). The two different CRYOs extracted were analyzed to compare the differences in the amount of fibrinogen recovered, clotting factors extracted, and clotting activity. RESULTS: The amount of fibrinogen in the CRYO extracted using the B-siphon method was similar to that obtained using the standard method (recovery of fibrinogen: B-siphon method: 71.2% vs. standard method: 61.0%). The amount of clotting XIII factor extracted using the B-siphon method was significantly lower than those extracted using the standard method. On the other hand, clotting II, V factors, and C1q esterase inhibitor not concentrated in CRYO content from the B-siphon method were significantly higher than that from the standard method. CONCLUSION: A new in-house CRYO preparation method was established by modifying a previously used thaw-siphon method. A coagulation factor-rich CRYO was extracted from plasma frozen at -40°C along with the first fraction of thawed plasma, without using a large-capacity refrigerated centrifuge for blood bags.


Subject(s)
Blood Coagulation Factors/analysis , Centrifugation/instrumentation , Cryopreservation/methods , Fibrinogen/analysis , Plasma/chemistry , Blood Coagulation Factors/metabolism , Chemical Precipitation , Complement C1 Inhibitor Protein/metabolism , Factor V/analysis , Factor VIII/analysis , Fibrinogen/metabolism , Humans , Indicators and Reagents/chemistry , Prothrombin/analysis
19.
Parasitology ; 148(7): 857-870, 2021 06.
Article in English | MEDLINE | ID: mdl-33729108

ABSTRACT

Angiostrongylus cantonensis is the main aetiological agent of eosinophilic meningoencephalitis in humans. Several outbreaks have been documented around the world, cementing its status as an emerging global public health concern. As a result, new strategies for the diagnosis, prophylaxis and treatment of cerebral angiostrongyliasis are urgently needed. In this study, we report on the de novo assembly of the A. cantonensis transcriptome, its full functional annotation and a reconstruction of complete metabolic pathways. All results are available at AngiostrongylusDB (http://angiostrongylus.lad.pucrs.br/admin/welcome). The aim of this study was to identify the active genes and metabolic pathways involved in the mechanisms of infection and survival inside Rattus norvegicus. Among 389 metabolic mapped pathways, the blood coagulation/antithrombin pathways of heparan sulphate/heparin are highlighted. Moreover, we identified genes codified to GP63 (leishmanolysin), CALR (calreticulin), ACE (peptidyl-dipeptidase A), myoglobin and vWD (von Willebrand factor type D domain protein) involved in the infection invasion and survival of the parasite. The large dataset of functional annotations provided and the full-length transcripts identified in this research may facilitate future functional genomics studies and provides a basis for the development of new techniques for the diagnosis, prevention and treatment of cerebral angiostrongyliasis.


Subject(s)
Antithrombins/metabolism , Blood Coagulation Factors/metabolism , Transcriptome , Angiostrongylus cantonensis , Animals , Female , Rats , Strongylida Infections
20.
Transfus Med ; 31(2): 136-141, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33686720

ABSTRACT

OBJECTIVES: Assessment of the impact of pooling five single-donor plasma (SDP) units to obtain six pathogen-reduced therapeutic plasma (PTP) units on standardisation and the retention of labile coagulation factors. BACKGROUND: SDP shows a high inter-donor variability with potential implications for the clinical treatment outcome. Additionally, there is still an existing risk for window-period transmissions of blood borne pathogens including newly emerging pathogens. METHODS/MATERIALS: Five ABO-identical SDP units were pooled, treated with the INTERTCEPT™ Blood System (Cerus Corporation, U.S.A.) and split into six PTP units which were frozen and thawed after 30 days. The variability in volume, labile coagulation factor retention and activity was assessed. RESULTS: The variability of volumes between the PTP units was reduced by 46% compared to SDP units. The variability in coagulation factor content between the PTP units was reduced by 63% compared to SDP units. Moderate, but significant losses of coagulation factors (except for vWF) were observed in PTPs compared to SDPs. CONCLUSION: The pooling of five SDP units to obtain six PTP units significantly increases product standardisation with potential implications for safety, economics as well as transfusion-transmitted pathogen safety, making it an interesting alternative to quarantine SDP (qSDP) and pathogen-reduced SDP.


Subject(s)
Blood Preservation/methods , Blood Preservation/standards , Furocoumarins/pharmacology , Photosensitizing Agents/pharmacology , Plasma , Ultraviolet Rays , Biomarkers/analysis , Biomarkers/blood , Blood Coagulation Factors/analysis , Blood Coagulation Factors/metabolism , Blood Safety/methods , Blood Safety/standards , Humans , Plasma/drug effects , Plasma/metabolism , Plasma/microbiology , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL