Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 232
Filtrer
1.
Article de Anglais | MEDLINE | ID: mdl-35718721

RÉSUMÉ

OBJECTIVE: To investigate the incorporation of the antifibrinolytic agent tranexamic acid (TA) during platelet-rich fibrin (PRF) formation to produce a robust fibrin agent with procoagulation properties. STUDY DESIGN: Blood from healthy volunteers was collected. Into 3 tubes, TA was immediately added in 1-mL, 0.4-mL, and 0.2-mL volumes, and the fourth tube was without additions. After PRF preparation, the clots were weighed in their raw (clot) and membrane forms. PRF physical properties were analyzed using a universal testing system (Instron). Protein and TA levels in the PRF were analyzed using a bicinchoninic acid assay and a ferric chloride assay, respectively. RESULTS: The addition of TA to PRF led to a robust weight compared with sham control. PRF weight was greater in females in all tested groups. The addition of TA also led to greater resilience to tears, especially at 1-mL TA addition to the blood. Furthermore, TA addition led to a greater value of total protein within the PRF and entrapment of TA in the PRF. CONCLUSIONS: Addition of TA to a PRF preparation leads to robust PRF with greater protein levels and the amalgamation of TA into the PRF. Such an agent may enhance the beneficial properties of PRF and attribute procoagulation properties to it.


Sujet(s)
Antifibrinolytiques , Hémostatiques , Fibrine riche en plaquettes , Acide tranéxamique , Antifibrinolytiques/métabolisme , Antifibrinolytiques/pharmacologie , Facteurs biologiques/métabolisme , Plaquettes , Centrifugation , Études de cohortes , Femelle , Fibrine/métabolisme , Humains , Mâle , Fibrine riche en plaquettes/métabolisme , Acide tranéxamique/métabolisme , Acide tranéxamique/pharmacologie
2.
Mol Immunol ; 142: 95-104, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-34973499

RÉSUMÉ

BACKGROUND: Excessive bradykinin (BK) generation from high molecular weight kininogen (HK) by plasma kallikrein (PK) due to lack of protease inhibition is central to the pathophysiology of hereditary angioedema (HAE). Inadequate protease inhibition may contribute to HAE through a number of plasma proteases including factor VII activating protease (FSAP) that can also cleave HK. OBJECTIVE: To investigate the interaction between FSAP and C1 inhibitor (C1Inh) and evaluate the potential role of FSAP in HAE with C1Inh deficiency. MATERIALS AND METHODS: Plasma samples from 20 persons with HAE types 1 or 2 in remission were studied and compared to healthy controls. We measured and compared antigenic FSAP levels, spontaneous FSAP activity, FSAP generation potential, activation of plasma pre-kallikrein (PPK) by FSAP, and the formation of FSAP-C1Inh and FSAP-alpha2-antiplasmin (FSAP-α2AP) complexes. Furthermore, we measured HK cleavage and PK activation after activation of endogenous pro-FSAP and after addition of exogenous FSAP. RESULTS: In plasma from HAE patients, there is increased basal FSAP activity compared to healthy volunteers. HAE plasma exhibits decreased formation of FSAP-C1Inh complexes and increased formation of FSAP-α2AP complexes in histone-activated plasma. Although exogenous FSAP can cleave HK in plasma, this was not seen when endogenous plasma pro-FSAP was activated with histones in either group. PK was also not activated by FSAP in plasma. CONCLUSION: In this study, we established that FSAP activity is increased and the pattern of FSAP-inhibitor complexes is altered in HAE patients. However, we did not find evidence suggesting that FSAP contributes directly to HAE attacks.


Sujet(s)
Angio-oedèmes héréditaires/physiopathologie , C1 Inhibiteur/génétique , Kininogène de haut poids moléculaire/métabolisme , Serine endopeptidases/métabolisme , Angio-oedèmes héréditaires/sang , Angio-oedèmes héréditaires/génétique , Antifibrinolytiques/métabolisme , Bradykinine/biosynthèse , Facteur VII/métabolisme , Humains , Kallicréines/sang , Kallicréines/métabolisme , Serine endopeptidases/génétique
3.
Cardiovasc Diabetol ; 20(1): 172, 2021 08 23.
Article de Anglais | MEDLINE | ID: mdl-34425843

RÉSUMÉ

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.


Sujet(s)
Antifibrinolytiques/métabolisme , Facteurs de la coagulation sanguine/métabolisme , COVID-19/complications , Adulte , Évolution de la maladie , Femelle , Humains , Mâle , Adulte d'âge moyen , SARS-CoV-2/pathogénicité , Syndrome de post-COVID-19
4.
Sci Rep ; 10(1): 8161, 2020 05 18.
Article de Anglais | MEDLINE | ID: mdl-32424181

RÉSUMÉ

Dietary odd-chain saturated fatty acids (OCFAs) are present in trace levels in dairy fat and some fish and plants. Higher circulating concentrations of OCFAs, pentadecanoic acid (C15:0) and heptadecanoic acid (C17:0), are associated with lower risks of cardiometabolic diseases, and higher dietary intake of OCFAs is associated with lower mortality. Population-wide circulating OCFA levels, however, have been declining over recent years. Here, we show C15:0 as an active dietary fatty acid that attenuates inflammation, anemia, dyslipidemia, and fibrosis in vivo, potentially by binding to key metabolic regulators and repairing mitochondrial function. This is the first demonstration of C15:0's direct role in attenuating multiple comorbidities using relevant physiological mechanisms at established circulating concentrations. Pairing our findings with evidence that (1) C15:0 is not readily made endogenously, (2) lower C15:0 dietary intake and blood concentrations are associated with higher mortality and a poorer physiological state, and (3) C15:0 has demonstrated activities and efficacy that parallel associated health benefits in humans, we propose C15:0 as a potential essential fatty acid. Further studies are needed to evaluate the potential impact of decades of reduced intake of OCFA-containing foods as contributors to C15:0 deficiencies and susceptibilities to chronic disease.


Sujet(s)
Acides gras/métabolisme , Inflammation/métabolisme , Animaux , Anti-inflammatoires/métabolisme , Antifibrinolytiques/métabolisme , Lignée cellulaire , Matières grasses alimentaires/métabolisme , Femelle , Humains , Inflammation/diétothérapie , Inflammation/prévention et contrôle , Foie , Mâle , Souris , Rat Sprague-Dawley
5.
J Med Chem ; 63(4): 1445-1472, 2020 02 27.
Article de Anglais | MEDLINE | ID: mdl-31658420

RÉSUMÉ

Hyperfibrinolytic situations can lead to life-threatening bleeding, especially during cardiac surgery. The approved antifibrinolytic agents such as tranexamic acid, ε-aminocaproic acid, 4-aminomethylbenzoic acid, and aprotinin were developed in the 1960s without the structural insight of their respective targets. Crystal structures of the main antifibrinolytic targets, the lysine binding sites on plasminogen's kringle domains, and plasmin's serine protease domain greatly contributed to the structure-based drug design of novel inhibitor classes. Two series of ligands targeting the lysine binding sites have been recently described, which are more potent than the most-widely used antifibrinolytic agent, tranexamic acid. Furthermore, four types of promising active site inhibitors of plasmin have been developed: tranexamic acid conjugates targeting the S1 pocket and primed sites, substrate-analogue linear homopiperidylalanine-containing 4-amidinobenzylamide derivatives, macrocyclic inhibitors addressing nonprimed binding regions, and bicyclic 14-mer SFTI-1 analogues blocking both, primed and nonprimed binding sites of plasmin. Furthermore, several allosteric plasmin inhibitors based on heparin mimetics have been developed.


Sujet(s)
Antifibrinolytiques/usage thérapeutique , Fibrinolyse/effets des médicaments et des substances chimiques , Hémorragie/traitement médicamenteux , Hémorragie/prévention et contrôle , Animaux , Antifibrinolytiques/composition chimique , Antifibrinolytiques/métabolisme , Domaine catalytique , Cristallographie aux rayons X , Fibrinolysine/composition chimique , Fibrinolysine/métabolisme , Humains , Ligands , Structure moléculaire , Plasminogène/composition chimique , Plasminogène/métabolisme , Liaison aux protéines , Domaines protéiques
6.
PLoS Negl Trop Dis ; 13(8): e0007660, 2019 08.
Article de Anglais | MEDLINE | ID: mdl-31449524

RÉSUMÉ

Ticks successfully feed and transmit pathogens by injecting pharmacological compounds in saliva to thwart host defenses. We have previously used LC-MS/MS to identify proteins that are present in saliva of unfed Amblyomma americanum ticks that were exposed to different hosts. Here we show that A. americanum serine protease inhibitor (serpin) 27 (AAS27) is an immunogenic saliva protein that is injected into the host within the first day of tick feeding and is an anti-inflammatory protein that might act by blocking plasmin and trypsin functions. Although AAS27 is injected into the host throughout tick feeding, qRT-PCR and western blotting analyses indicate that the respective transcript and protein are present in high amounts within the first 24 h of tick feeding. Biochemical screening of Pichia pastoris-expressed recombinant (r) AAS27 against mammalian proteases related to host defense shows it is an inhibitor of trypsin and plasmin, with stoichiometry of inhibition indices of 3.5 and 3.8, respectively. Consistent with typical inhibitory serpins, rAAS27 formed heat- and SDS-stable irreversible complexes with both proteases. We further demonstrate that rAAS27 inhibits trypsin with ka of 6.46 ± 1.24 x 104 M-1 s-1, comparable to serpins of other tick species. We show that native AAS27 is part of the repertoire of proteins responsible for the inhibitory activity against trypsin in crude tick saliva. AAS27 is likely utilized by the tick to evade the hosts inflammation defense since rAAS27 blocks both formalin and compound 48/80-induced inflammation in rats. Tick immune sera of rabbits that had acquired resistance against tick feeding following repeated infestations with A. americanum or Ixodes scapularis ticks reacts with rAAS27. Of significant interest, antibody to rAAS27 blocks this serpin inhibitory functions. Taken together, we conclude that AAS27 is an anti-inflammatory protein secreted into the host during feeding and may represent a potential candidate for development of an anti-tick vaccine.


Sujet(s)
Anti-inflammatoires/métabolisme , Protéines d'arthropode/métabolisme , Échappement immunitaire , Ixodidae/pathogénicité , Serpines/métabolisme , Animaux , Antifibrinolytiques/métabolisme , Transport des protéines , Lapins , Rats , Inhibiteurs trypsiques/métabolisme
7.
Pol Merkur Lekarski ; 44(261): 135-138, 2018 Mar 27.
Article de Polonais | MEDLINE | ID: mdl-29601563

RÉSUMÉ

The implementation of treatment in patients with sarcoidosis (SA) must be associated with the certainty of diagnosis, which is difficult due to the lack of unambiguous criteria. Finding the presence of noncaseating granulomas in bioptic material is not always indicative of SA. The main point of SA's diagnosis is the level of its activity, because only patients in the active stage should be qualified for treatment. In therapy, glucocorticosteroids or second-line drugs - methotrexate or azathioprine are still recommended. Introduced monoclonal antibodies (infliximab, etanercept, adaluimumab, golimumab, rituximab), tested for efficacy in other pathologies associated with the formation of granulomas, have a limited application in patients with SA. In contrast, anti-fibrotics (pirfenidone and nintedanib) are in clinical trials. The latest method of controlling the fibrosis of the parenchyma in the course of SA is the use of mesenchymal cells obtained from umbilical cord blood. Preliminary results indicate a real possibility of using this therapy in patients with SA.


Sujet(s)
Sarcoïdose/diagnostic , Sarcoïdose/traitement médicamenteux , Hormones corticosurrénaliennes/usage thérapeutique , Anticorps monoclonaux humanisés/usage thérapeutique , Antifibrinolytiques/métabolisme , Azathioprine/usage thérapeutique , Biothérapie , Diagnostic différentiel , Humains , Méthotrexate/métabolisme , Guides de bonnes pratiques cliniques comme sujet , Sarcoïdose/thérapie , Transplantation de cellules souches
8.
Arch Physiol Biochem ; 124(2): 156-166, 2018 May.
Article de Anglais | MEDLINE | ID: mdl-28857634

RÉSUMÉ

The current study aimed to investigate the effect of Citrullus colocynthis (C. colocynthis) hydro-alcoholic extract on blood haemostasis in control and high-fat diet (HFD) induced obese rats. In control rats, the extract significantly enhanced bleeding time and plasma levels of tPA and significantly decreased plasma levels PAI-1 and serum levels of thromboxane B2 leading to inhibition of platelets aggregation. In HFD induced obese rats, similar effects were seen and the extract was also able to reverse HFD induced increases in fibrinogen and VWF. Searching for the mechanism, C. colocynthis acts by (1) inhibiting of food intake, (2) inhibiting the activity of pancreatic lipase, (3) decreasing levels of TNF-α and IL-6 and (4) decreasing circulatory levels of the prothrombotic adipokine, leptin and enhanced circulatory levels of the antithrombic adipokines and adiopnectin. In conclusion, C. colocynthis has antiplatelets and profibrinolytic activity in both control and HFD induced obese rats.


Sujet(s)
Agents antiobésité/usage thérapeutique , Antifibrinolytiques/usage thérapeutique , Citrullus colocynthis/composition chimique , Compléments alimentaires , Obésité/prévention et contrôle , Extraits de plantes/usage thérapeutique , Antiagrégants plaquettaires/usage thérapeutique , Animaux , Agents antiobésité/effets indésirables , Agents antiobésité/métabolisme , Antifibrinolytiques/effets indésirables , Antifibrinolytiques/métabolisme , Anorexigènes/effets indésirables , Anorexigènes/métabolisme , Anorexigènes/usage thérapeutique , Marqueurs biologiques/analyse , Marqueurs biologiques/sang , Marqueurs biologiques/urine , Temps de saignement , Alimentation riche en graisse/effets indésirables , Compléments alimentaires/effets indésirables , Fèces/composition chimique , Fibrinolyse , Fruit/composition chimique , Métabolisme lipidique , Mâle , Obésité/sang , Obésité/étiologie , Obésité/métabolisme , Extraits de plantes/effets indésirables , Extraits de plantes/métabolisme , Inhibiteur-1 d'activateur du plasminogène/sang , Agrégation plaquettaire , Antiagrégants plaquettaires/effets indésirables , Antiagrégants plaquettaires/métabolisme , Rat Sprague-Dawley , Thromboxane B2/sang , Activateur tissulaire du plasminogène/sang
9.
Anesth Analg ; 126(2): 439-442, 2018 02.
Article de Anglais | MEDLINE | ID: mdl-28678070

RÉSUMÉ

Prior studies have suggested that the antifibrinolytic drug aprotinin increases the infarct size after ischemia and reperfusion (I/R) and attenuates the effect of ischemic preconditioning (IPC). Aprotinin was replaced by tranexamic acid (TXA) in clinical practice. Here, we investigated whether TXA influences I/R injury and/or cardioprotection initiated by IPC and/or remote ischemic preconditioning (RIPC). Anesthetized male Wistar rats were randomized to 6 groups. Control animals were not further treated. Administration of TXA was combined with and without IPC and RIPC. Estimated treatment effect was 20%. Compared to control group (56% ± 11%), IPC reduced infarct size by 46% (30% ± 6%; mean difference, 26%; 95% confidence interval, 19-33; P < .0001), and RIPC reduced infarct size by 29% (40% ± 8%; mean difference, 16%; 95% confidence interval, 9-24; P < .011). Additional application of TXA had no effect on I/R injury and cardioprotection by IPC or RIPC. TXA does not abolish infarct size reduction by IPC or RIPC.


Sujet(s)
Antifibrinolytiques/administration et posologie , Préconditionnement ischémique myocardique/méthodes , Ischémie myocardique/prévention et contrôle , Acide tranéxamique/administration et posologie , Animaux , Antifibrinolytiques/métabolisme , Mâle , Ischémie myocardique/métabolisme , Ischémie myocardique/anatomopathologie , Rats , Rat Wistar
10.
Article de Anglais | MEDLINE | ID: mdl-28917645

RÉSUMÉ

Bee venom contains a variety of peptide constituents, including low-molecular-weight protease inhibitors. While the putative low-molecular-weight serine protease inhibitor Api m 6 containing a trypsin inhibitor-like cysteine-rich domain was identified from honeybee (Apis mellifera) venom, no anti-fibrinolytic or anti-microbial roles for this inhibitor have been elucidated. In this study, we identified an Asiatic honeybee (A. cerana) venom serine protease inhibitor (AcVSPI) that was shown to act as a microbial serine protease inhibitor and plasmin inhibitor. AcVSPI was found to consist of a trypsin inhibitor-like domain that displays ten cysteine residues. Interestingly, the AcVSPI peptide sequence exhibited high similarity to the putative low-molecular-weight serine protease inhibitor Api m 6, which suggests that AcVSPI is an allergen Api m 6-like peptide. Recombinant AcVSPI was expressed in baculovirus-infected insect cells, and it demonstrated inhibitory activity against trypsin, but not chymotrypsin. Additionally, AcVSPI has inhibitory effects against plasmin and microbial serine proteases; however, it does not have any detectable inhibitory effects on thrombin or elastase. Consistent with these inhibitory effects, AcVSPI inhibited the plasmin-mediated degradation of fibrin to fibrin degradation products. AcVSPI also bound to bacterial and fungal surfaces and exhibited anti-microbial activity against fungi as well as gram-positive and gram-negative bacteria. These findings demonstrate the anti-fibrinolytic and anti-microbial roles of AcVSPI as a serine protease inhibitor.


Sujet(s)
Antibactériens/pharmacologie , Venins d'abeille/métabolisme , Abeilles/métabolisme , Inhibiteurs de la sérine protéinase/pharmacologie , Animaux , Antibactériens/métabolisme , Antifibrinolytiques/métabolisme , Venins d'abeille/composition chimique , Clonage moléculaire , ADN complémentaire , Régulation de l'expression des gènes/physiologie , Protéines d'insecte/génétique , Protéines d'insecte/métabolisme , Inhibiteurs de la sérine protéinase/composition chimique , Inhibiteurs de la sérine protéinase/génétique , Inhibiteurs de la sérine protéinase/métabolisme
11.
Biomed Res Int ; 2017: 1209676, 2017.
Article de Anglais | MEDLINE | ID: mdl-28761875

RÉSUMÉ

The inhibitory effect of coagulation factor XIII (FXIII) on fibrinolysis has been studied for at least 50 years. Our insight into the underlying mechanisms has improved considerably, aided in particular by the discovery that activated FXIII cross-links α2-antiplasmin (α2AP) to fibrin. In this review, the most important effects of different cross-linking reactions on fibrinolysis are summarized. A distinction is made between fibrin-fibrin cross-links studied in purified systems and fibrin-α2AP cross-links studied in plasma or whole blood systems. While the formation of γ chain dimers in fibrin does not affect clot lysis, the formation of α chain polymers has a weak inhibitory effect. Only strong cross-linking of fibrin, associated with high molecular weight α chain polymers and/or γ chain multimers, results in a moderate inhibition fibrinolysis. The formation of fibrin-α2AP cross-links has only a weak effect on clot lysis, but this effect becomes strong when clot retraction occurs. Under these conditions, FXIII prevents α2AP being expelled from the clot and makes the clot relatively resistant to degradation by plasmin.


Sujet(s)
Facteur XIII/métabolisme , Fibrinolyse/physiologie , Antifibrinolytiques/métabolisme , Coagulation sanguine/physiologie , Fibrine/métabolisme , Humains
12.
J Chem Inf Model ; 57(7): 1703-1714, 2017 07 24.
Article de Anglais | MEDLINE | ID: mdl-28653850

RÉSUMÉ

Inhibition of plasmin has been found to effectively reduce fibrinolysis and to avoid hemorrhage. This can be achieved by addressing its kringle 1 domain with the known drug and lysine analogue tranexamic acid. Guided by shape similarities toward a previously discovered lead compound, 5-(4-piperidyl)isoxazol-3-ol, a set of 16 structurally similar compounds was assembled and investigated. Successfully, in vitro measurements revealed one compound, 5-(4-piperidyl)isothiazol-3-ol, superior in potency compared to the initial lead. Furthermore, a strikingly high correlation (R2 = 0.93) between anti-fibrinolytic activity and kringle 1 binding affinity provided strong support for the hypothesized inhibition mechanism, as well as revealing opportunities to fine-tune biological effects through minor structural modifications. Several different ligand-based (Freeform, shape, and electrostatic-based similarities) and structure-based methods (e.g., Posit, MM/GBSA, FEP+) were used to retrospectively predict the binding affinities. A combined method, molecular alignment using Posit and scoring with Tcombo, lead to the highest coefficient of determination (R2 = 0.6).


Sujet(s)
Antifibrinolytiques/composition chimique , Antifibrinolytiques/pharmacologie , Découverte de médicament , Fibrinolysine/antagonistes et inhibiteurs , Isoxazoles/composition chimique , Isoxazoles/pharmacologie , Pipéridines/composition chimique , Pipéridines/pharmacologie , Antifibrinolytiques/métabolisme , Fibrinolysine/composition chimique , Fibrinolysine/métabolisme , Isoxazoles/métabolisme , Simulation de docking moléculaire , Pipéridines/métabolisme , Domaines protéiques , Relation quantitative structure-activité , Thermodynamique
13.
Mol Nutr Food Res ; 61(4)2017 04.
Article de Anglais | MEDLINE | ID: mdl-27983763

RÉSUMÉ

SCOPE: Myricetin is found in most berries, vegetables, and various medicinal herbs, which has been reported to possess various bio-activities. However, the role of myricetin on liver fibrosis remains to be elucidated. METHODS AND RESULTS: Hepatic stellate cell (HSC) line CFSC-8B was stimulated by transforming growth factor ß1 (TGF-ß1) or platelet-derived growth factor BB (PDGF-BB) to induce liver fibrosis in vitro. The results showed that myricetin significantly ameliorated TGF-ß1- or PDGF-BB-induced HSCs activation, cell migration, and extracellular matrix production; blocked TGF-ß1-induced phosphorylation of Smad2, P38, extracellular signal-regulated kinase (ERK), and protein kinase B (Akt); and downregulated PDGF-BB stimulated phosphorylation of extracellular signal-regulated kinase and Akt in HSCs in a dose-dependent manner. Meanwhile, the carbon tetrachloride (CCl4 ) induced mouse model has been used to study antifibrosis role of myricetin in vivo. Our data demonstrated that myricetin suppressed α-smooth muscle actin and collagen type I deposition and blocked phosphorylation of Smad2, mitogen-activated protein kinases, and Akt in CCl4 -treated mice. CONCLUSION: Myricetin inhibits the activation of HSCs and ameliorates CCl4 -induced liver fibrosis in mice and may serve as a potential therapeutic agent in the treatment of liver fibrosis.


Sujet(s)
Tétrachloro-méthane/effets indésirables , Flavonoïdes/pharmacologie , Facteur de croissance dérivé des plaquettes/métabolisme , Facteur de croissance transformant bêta-1/métabolisme , Animaux , Antifibrinolytiques/métabolisme , Bécaplermine , Mouvement cellulaire/effets des médicaments et des substances chimiques , Collagène de type I/métabolisme , Modèles animaux de maladie humaine , Extracellular Signal-Regulated MAP Kinases/métabolisme , Cellules étoilées du foie/métabolisme , Cirrhose du foie/induit chimiquement , Cirrhose du foie/traitement médicamenteux , Souris , Phosphorylation , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-sis/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Facteur de croissance transformant bêta/métabolisme
14.
Perfusion ; 32(3): 226-229, 2017 Apr.
Article de Anglais | MEDLINE | ID: mdl-27815558

RÉSUMÉ

OBJECTIVE: To assess the in vitro effects of drug sequestration in extracorporeal membrane oxygenation (ECMO) on ϵ-aminocaproic acid (EACA) concentrations. METHODS AND DESIGN: This in vitro study will determine changes in EACA concentration over time in ECMO circuits. A pediatric dose of 2,500 mg was administered to whole expired blood in the simulated pediatric ECMO circuit. Blood samples were collected at 0, 30, 60, 360 and 1440-minute intervals after initial administration equilibration from three different sites of the circuit: pre-oxygenator (PRE), post-oxygenator (POST) and PVC tubing (PVC) to determine the predominant site of drug loss. The circuit was maintained for two consecutive days with a re-dose at 24 hours to establish a comparison between unsaturated (New) and saturated (Old) oxygenator membranes. Comparisons between sample sites, sample times and New versus Old membranes were statistically analyzed by a linear mixed-effects model with significance defined as a p-value <0.05. RESULTS: There were no significant differences in EACA concentration with respect to sample site, with PRE and POST samples demonstrating respective mean differences of 0.30 mg/ml and 0.34 mg/ml as compared to PVC, resulting in non-significant p-values of 0.373 [95% CI (-0.37, 0.98)] and 0.324 [95% CI (-0.34, 1.01)], respectively. The comparison of New vs. Old ECMO circuits resulted in non-significant changes from baseline, with a mean difference of 0.50 mg/ml, 95% CI (-0.65, 1.65), p=0.315. CONCLUSION: The findings of this study did not show any significant changes in drug concentration that can be attributed to sequestration within the ECMO circuit. Mean concentrations between ECMO circuit sample sites did not differ significantly. Comparison between New and Old circuits also did not differ significantly in the change from baseline concentration over time. Sequestration within ECMO circuits appears not to be a considerable factor for EACA administration.


Sujet(s)
Acide 6-amino-caproïque/analyse , Antifibrinolytiques/analyse , Oxygénation extracorporelle sur oxygénateur à membrane/instrumentation , Acide 6-amino-caproïque/métabolisme , Antifibrinolytiques/métabolisme , Humains , Oxygénateurs à membrane
15.
Blood Coagul Fibrinolysis ; 28(6): 431-437, 2017 Sep.
Article de Anglais | MEDLINE | ID: mdl-27898513

RÉSUMÉ

: The clinical limitations of unfractionated heparin (UFH) and low molecular weight heparin (LMWH) led to the development of an antithrombin-heparin covalent complex (ATH), which displays superior anticoagulant abilities compared with UFH. A recent study investigating its interaction with fibrinolysis showed that ATH inhibited free and fibrin bound plasmin and decreased plasmin generation on fibrin clots. These studies were conducted using purified components and did not elucidate the interaction of ATH with plasmin in the presence of its natural inhibitors α2-antiplasmin (α2-AP) and α2-macroglobulin (α2-M). The aim of this study was to determine the effects of ATH, UFH, and LMWH on plasmin generation in plasma, under more physiological conditions. Plasmin generation in plasma in the absence and presence of anticoagulants was initiated by tissue plasminogen activator and soluble fibrin fragments, and plasmin and plasmin-α2-M complexes generated over time were quantified chromogenically. Generation of plasmin-α2-AP complexes and consumption of plasminogen were quantified by ELISA. Plasmin generation was decreased in the presence of UFH and ATH, whereas LMWH had no effect. Neither plasminogen consumption nor generation of plasmin-α2-AP complexes were affected by UFH or ATH. However, plasmin-α2-M complexes were slightly reduced by ATH suggesting that ATH may be able to compete with α2-M for plasmin. Plasmin generation may be mildly inhibited by heparin-based anticoagulants; however, heparin-catalyzed antithrombin activity is not a major inhibitor of plasmin, as compared to its natural inhibitors α2-AP and α2-M. This adds to our understanding of ATH mechanisms of action and aids in its development for clinical use.


Sujet(s)
Antithrombiniques/pharmacologie , Fibrinolysine/antagonistes et inhibiteurs , Héparine bas poids moléculaire/pharmacologie , Héparine/pharmacologie , Antifibrinolytiques/métabolisme , Antithrombiniques/composition chimique , Fibrinolysine/métabolisme , Héparine/composition chimique , Humains , Complexes multiprotéiques/composition chimique , Complexes multiprotéiques/pharmacologie , Plasma sanguin/métabolisme , alpha 2-Macroglobulines associées à la grossesse/métabolisme
16.
Dev Comp Immunol ; 63: 27-35, 2016 10.
Article de Anglais | MEDLINE | ID: mdl-27208884

RÉSUMÉ

Bee venom contains a variety of peptide constituents that have various biological, toxicological, and pharmacological actions. However, the biological actions of secapin, a venom peptide in bee venom, remain largely unknown. Here, we provide the evidence that Asiatic honeybee (Apis cerana) secapin (AcSecapin-1) exhibits anti-fibrinolytic, anti-elastolytic, and anti-microbial activities. The recombinant mature AcSecapin-1 peptide was expressed in baculovirus-infected insect cells. AcSecapin-1 functions as a serine protease inhibitor-like peptide that has inhibitory effects against plasmin, elastases, microbial serine proteases, trypsin, and chymotrypsin. Consistent with these functions, AcSecapin-1 inhibited the plasmin-mediated degradation of fibrin to fibrin degradation products, thus indicating the role of AcSecapin-1 as an anti-fibrinolytic agent. AcSecapin-1 also inhibited both human neutrophil and porcine pancreatic elastases. Furthermore, AcSecapin-1 bound to bacterial and fungal surfaces and exhibited anti-microbial activity against fungi and gram-positive and gram-negative bacteria. Taken together, our data demonstrated that the bee venom peptide secapin has multifunctional roles as an anti-fibrinolytic agent during fibrinolysis and an anti-microbial agent in the innate immune response.


Sujet(s)
Venins d'abeille/métabolisme , Abeilles/immunologie , Immunité innée , Protéines d'insecte/métabolisme , Animaux , Anti-infectieux/métabolisme , Antifibrinolytiques/métabolisme , Baculoviridae/génétique , Venins d'abeille/génétique , Lignée cellulaire , Humains , Protéines d'insecte/génétique , Leukocyte elastase/antagonistes et inhibiteurs , Pancreatic elastase/antagonistes et inhibiteurs , Suidae
17.
Transfusion ; 56 Suppl 2: S110-4, 2016 04.
Article de Anglais | MEDLINE | ID: mdl-27100746

RÉSUMÉ

Postinjury fibrinolysis can manifest as three distinguishable phenotypes: 1) hyperfibrinolysis, 2) physiologic, and 3) hypofibrinolysis (shutdown). Hyperfibrinolysis is associated with uncontrolled bleeding due to clot dissolution; whereas, fibrinolysis shutdown is associated with organ dysfunction due to microvascular occlusion. The incidence of fibrinolysis phenotypes at hospital arrival in severely injured patients is: 1) hyperfibrinolysis 18%, physiologic 18%, and shutdown 64%. The mechanisms responsible for dysregulated fibrinolysis following injury remain uncertain. Animal work suggests hypoperfusion promotes fibrinolysis, while tissue injury inhibits fibrinolysis. Clinical experience is consistent with these observations. The predominant mediator of postinjury hyperfibrinolysis appears to be tissue plasminogen activator (tPA) released from ischemic endothelium. The effects of tPA are accentuated by impaired hepatic clearance. Fibrinolysis shutdown, on the other hand, may occur from inhibition of circulating tPA, enhanced clot strength impairing the binding of tPA and plasminogen to fibrin, or the inhibition of plasmin. Plasminogen activator inhibitor -1 (PAI-1) binding of circulating tPA appears to be a major mechanism for postinjury shutdown. The sources of PAI-1 include endothelium, platelets, and organ parenchyma. The laboratory identification of fibrinolysis phenotype, at this moment, is best determined with viscoelastic hemostatic assays (TEG, ROTEM). While D-dimer and plasmin antiplasmin (PAP) levels corroborate fibrinolysis, they do not provide real-time assessment of the circulating blood capacity. Our clinical studies indicate that fibrinolysis is a very dynamic process and our experimental work suggests plasma first resuscitation reverses hyperfibrinolysis. Collectively, we believe recent clinical and experimental work suggest antifibrinolytic therapy should be employed selectively in the acutely injured patient, and optimally guided by TEG or ROTEM.


Sujet(s)
Fibrinolyse/effets des médicaments et des substances chimiques , Acide tranéxamique/usage thérapeutique , Plaies et blessures/traitement médicamenteux , Plaies et blessures/métabolisme , Animaux , Antifibrinolytiques/métabolisme , Antifibrinolytiques/usage thérapeutique , Fibrine/métabolisme , Humains , Phénotype , Plasminogène/métabolisme , Inhibiteur-1 d'activateur du plasminogène/métabolisme , Activateur tissulaire du plasminogène/métabolisme
18.
Medicine (Baltimore) ; 95(10): e2895, 2016 Mar.
Article de Anglais | MEDLINE | ID: mdl-26962786

RÉSUMÉ

Educational advice is often given to patients starting treatment with vitamin K Antagonists (VKAs). A great emphasis is made on nutritional information. Common belief is that dietary vitamin K intake could counteract the anticoagulant effect by VKAs and for many years, patients have been discouraged to consume vitamin-K-rich foods, such as green leafy vegetables.The objective of this study is to summarize the current evidence supporting the putative interaction between dietary vitamin K intake and changes in INR with the VKAs.Data sources are MEDLINE via PubMed and Cochrane database.All clinical studies investigating the relationship between dietary vitamin K and measures of anticoagulation were included. We excluded all studies of supplementation of vitamin K alone.We performed a systematic review of the literature up to October 2015, searching for a combination of "food," "diet," "vitamin K," "phylloquinone," "warfarin," "INR," "coagulation," and "anticoagulant."Two dietary interventional trials and 9 observational studies were included. We found conflicting evidence on the effect of dietary intake of vitamin K on coagulation response. Some studies found a negative correlation between vitamin K intake and INR changes, while others suggested that a minimum amount of vitamin K is required to maintain an adequate anticoagulation. Median dietary intake of vitamin K1 ranged from 76 to 217 µg/day among studies, and an effect on coagulation may be detected only for high amount of vitamin intake (>150 µg/day).Most studies included patients with various indications for VKAs therapy, such as atrial fibrillation, prosthetic heart valves, and venous thromboembolism. Thus, INR target was dishomogeneous and no subanalyses for specific populations or different anticoagulants were conducted. Measures used to evaluate anticoagulation stability were variable.The available evidence does not support current advice to modify dietary habits when starting therapy with VKAs. Restriction of dietary vitamin K intake does not seem to be a valid strategy to improve anticoagulation quality with VKAs. It would be, perhaps, more relevant to maintain stable dietary habit, avoiding wide changes in the intake of vitamin K.


Sujet(s)
Coagulation sanguine/effets des médicaments et des substances chimiques , Compléments alimentaires , Thromboembolie , Vitamine K , Antifibrinolytiques/métabolisme , Antifibrinolytiques/pharmacologie , Humains , Rapport international normalisé , Besoins nutritifs , Thromboembolie/sang , Thromboembolie/prévention et contrôle , Vitamine K/antagonistes et inhibiteurs , Vitamine K/métabolisme , Vitamine K/pharmacologie
20.
Bone ; 79: 233-41, 2015 Oct.
Article de Anglais | MEDLINE | ID: mdl-26094563

RÉSUMÉ

The mechanism of postmenopausal osteoporosis is not fully understood. α2-Antiplasmin (α2-AP) is the primary inhibitor of plasmin in the fibrinolytic system, but is known to have activities beyond fibrinolysis. However, its role in bone metabolism and the pathogenesis of osteoporosis remains unknown. In the current study, we therefore examined the effects of α2-AP deficiency on ovariectomy (OVX)-induced bone loss by using wild-type and α2-AP-deficient mice. Quantitative computed tomography analysis revealed that α2-AP deficiency blunted OVX-induced trabecular bone loss in mice. Moreover, α2-AP deficiency significantly blunted serum levels of bone-specific alkaline phosphatase, cross-linked C-telopeptide of type I collagen, and interleukin (IL)-1ß elevated by OVX. α2-AP treatment elevated the levels of IL-1ß and tumor necrosis factor (TNF)-α mRNA in RAW 264.7 cells, although it suppressed osteoclast formation induced by receptor activator of nuclear factor-κB ligand. α2-AP treatment activated ERK1/2 and p38 MAP kinase pathways in RAW 264.7 cells, and these MAP kinase inhibitors antagonized the levels of IL-1ß mRNA elevated by α2-AP. The data demonstrate that α2-AP is linked to bone loss due to OVX, through a mechanism that depends in part on the production of IL-1ß and TNF-α in monocytes.


Sujet(s)
Antifibrinolytiques/métabolisme , Ostéoporose post-ménopausique/métabolisme , Transduction du signal/physiologie , Animaux , Technique de Western , Os et tissu osseux/métabolisme , Modèles animaux de maladie humaine , Femelle , Humains , Interleukine-1 bêta/métabolisme , Souris , Souris de lignée C57BL , Souris knockout , Ovariectomie , Réaction de polymérisation en chaine en temps réel , Tomodensitométrie , Facteur de nécrose tumorale alpha/métabolisme , p38 Mitogen-Activated Protein Kinases/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...