Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.121
Filter
1.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Article in English | MEDLINE | ID: mdl-33443167

ABSTRACT

The blood-clotting protein fibrinogen has been implicated in host defense following Staphylococcus aureus infection, but precise mechanisms of host protection and pathogen clearance remain undefined. Peritonitis caused by staphylococci species is a complication for patients with cirrhosis, indwelling catheters, or undergoing peritoneal dialysis. Here, we sought to characterize possible mechanisms of fibrin(ogen)-mediated antimicrobial responses. Wild-type (WT) (Fib+) mice rapidly cleared S. aureus following intraperitoneal infection with elimination of ∼99% of an initial inoculum within 15 min. In contrast, fibrinogen-deficient (Fib-) mice failed to clear the microbe. The genotype-dependent disparity in early clearance resulted in a significant difference in host mortality whereby Fib+ mice uniformly survived whereas Fib- mice exhibited high mortality rates within 24 h. Fibrin(ogen)-mediated bacterial clearance was dependent on (pro)thrombin procoagulant function, supporting a suspected role for fibrin polymerization in this mechanism. Unexpectedly, the primary host initiator of coagulation, tissue factor, was found to be dispensable for this antimicrobial activity. Rather, the bacteria-derived prothrombin activator vWbp was identified as the source of the thrombin-generating potential underlying fibrin(ogen)-dependent bacterial clearance. Mice failed to eliminate S. aureus deficient in vWbp, but clearance of these same microbes in WT mice was restored if active thrombin was administered to the peritoneal cavity. These studies establish that the thrombin/fibrinogen axis is fundamental to host antimicrobial defense, offer a possible explanation for the clinical observation that coagulase-negative staphylococci are a highly prominent infectious agent in peritonitis, and suggest caution against anticoagulants in individuals susceptible to peritoneal infections.


Subject(s)
Fibrinogen/metabolism , Peritonitis/metabolism , Prothrombin/metabolism , Animals , Anti-Bacterial Agents/metabolism , Anti-Infective Agents/metabolism , Blood Coagulation , Coagulase/metabolism , Female , Fibrin/metabolism , Fibrinogen/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Staphylococcal Infections/microbiology , Staphylococcus aureus/metabolism , Staphylococcus aureus/pathogenicity , Thromboplastin
2.
J Bacteriol ; 205(4): e0045122, 2023 04 25.
Article in English | MEDLINE | ID: mdl-36951588

ABSTRACT

Considerable progress has been made toward elucidating the mechanism of Staphylococcus aureus aggregation in synovial fluid. In this study, aggregate morphology was assessed following incubation under several simulated postsurgical joint conditions. Using fluorescently labeled synovial fluid polymers, we show that aggregation occurs through two distinct mechanisms: (i) direct bridging between S. aureus cells and host fibrinogen and (ii) an entropy-driven depletion mechanism facilitated by hyaluronic acid and albumin. By screening surface adhesin-deficient mutants (clfA, clfB, fnbB, and fnbA), we identified the primary genetic determinant of aggregation in synovial fluid to be clumping factor A. To characterize this bridging interaction, we employed an atomic force microscopy-based approach to quantify the binding affinity of either wild-type S. aureus or the adhesin mutant to immobilized fibrinogen. Surprisingly, we found there to be cell-to-cell variability in the binding strength of the bacteria for immobilized fibrinogen. Superhigh-resolution microscopy imaging revealed that fibrinogen binding to the cell wall is heterogeneously distributed at both the single cell and population levels. Finally, we assessed the antibiotic tolerance of various aggregate morphologies arising from newly deciphered mechanisms of polymer-mediated synovial fluid-induced aggregation. The formation of macroscopic aggregates under shear was highly tolerant of gentamicin, while smaller aggregates, formed under static conditions, were susceptible. We hypothesize that aggregate formation in the joint cavity, in combination with shear, is mediated by both polymer-mediated aggregation mechanisms, with depletion forces enhancing the stability of essential bridging interactions. IMPORTANCE The formation of a bacterial biofilm in the postsurgical joint environment significantly complicates the resolution of an infection. To form a resilient biofilm, incoming bacteria must first survive the initial invasion of the joint space. We previously found that synovial fluid induces the formation of Staphylococcus aureus aggregates, which may provide rapid protection during the early stages of infection. The state of the host joint environment, including the presence of fluid flow and fluctuating abundance of synovial fluid polymers, determines the rate and size of aggregate formation. By expanding on our knowledge of the mechanism and pathogenic implications of synovial fluid-induced aggregation, we hope to contribute insights for the development of novel methods of prevention and therapeutic intervention.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Humans , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Biofilms , Staphylococcal Infections/microbiology , Fibrinogen/metabolism , Fibrinogen/pharmacology
3.
Mol Syst Biol ; 18(2): e10629, 2022 02.
Article in English | MEDLINE | ID: mdl-35156780

ABSTRACT

Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) is a chloride and bicarbonate channel in secretory epithelia with a critical role in maintaining fluid homeostasis. Mutations in CFTR are associated with Cystic Fibrosis (CF), the most common lethal autosomal recessive disorder in Caucasians. While remarkable treatment advances have been made recently in the form of modulator drugs directly rescuing CFTR dysfunction, there is still considerable scope for improvement of therapeutic effectiveness. Here, we report the application of a high-throughput screening variant of the Mammalian Membrane Two-Hybrid (MaMTH-HTS) to map the protein-protein interactions of wild-type (wt) and mutant CFTR (F508del), in an effort to better understand CF cellular effects and identify new drug targets for patient-specific treatments. Combined with functional validation in multiple disease models, we have uncovered candidate proteins with potential roles in CFTR function/CF pathophysiology, including Fibrinogen Like 2 (FGL2), which we demonstrate in patient-derived intestinal organoids has a significant effect on CFTR functional expression.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator , Cystic Fibrosis , Animals , Cell Membrane/metabolism , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Fibrinogen/genetics , Fibrinogen/metabolism , Fibrinogen/pharmacology , High-Throughput Screening Assays , Humans , Mammals , Mutation
4.
Haemophilia ; 29(3): 836-843, 2023 May.
Article in English | MEDLINE | ID: mdl-36757142

ABSTRACT

INTRODUCTION: Hereditary hypofibrinogenemia is a rare fibrinogen disorder characterised by decreased levels of fibrinogen. Pregnant women with hypofibrinogenemia are at risk of adverse obstetrical outcomes, depending on the fibrinogen level. AIM: We investigated how the physiological changes of hemostasis throughout the pregnancy impact the hemostatic balance in a woman with hereditary mild hypofibrinogenemia. METHODS: Fibrin clot properties were analyzed by turbidimetry and scanning electron microscopy, clot weight and red blood cells retention were measured by whole clot contraction, and in vitro thrombin generation was assessed by calibrated automated thrombogram and ex vivo by TAT. RESULTS: Throughout the pregnancy, the fibrinogen levels increased reaching normal values in the third trimester (activity 3.1 g/L, antigen 3.2 g/L). In parallel, the fibrin polymerisation increased, the fibrinolysis decreased, the fibrin clot network became denser with thicker fibrin fibers, and the fibrin clot weight and red blood cells retention increased, reaching control's value at the third trimester. Similarly, in vitro and ex vitro thrombin generation increased, reaching maximum values at the delivery. CONCLUSION: In this case of hereditary mild hypofibrinogenemia we observed a physiological increase of fibrinogen and thrombin generation. Future studies should focus on moderate and severe hypofibrinogenemia, to assess fibrinogen variation and the overall impact of increased TG on the hemostasis balance.


Subject(s)
Afibrinogenemia , Hemostatics , Thrombosis , Pregnancy , Humans , Female , Blood Coagulation , Thrombin , Afibrinogenemia/genetics , Fibrinolysis , Fibrin , Hemostatics/pharmacology , Fibrinogen/pharmacology
5.
Cell Commun Signal ; 21(1): 278, 2023 10 10.
Article in English | MEDLINE | ID: mdl-37817162

ABSTRACT

BACKGROUND: Abnormal platelet activation is a key factor in the occurrence and development of thrombotic diseases. However, the physiological mechanisms that underlie platelet homeostasis remain unclear. Oleic acid, one of the most abundant lipids in the human diet, has potential antithrombotic effects. This study aimed to investigate the effects of oleic acid on platelet activation and thrombosis. METHODS: Platelet aggregation, ATP release, and fibrinogen spread were evaluated to determine the role of oleic acid in platelet activation. A ferric chloride-induced carotid injury model was used to establish the effect of oleic acid on thrombus formation in vivo. Western blotting analysis and transfection experiments were performed to determine the mechanisms involved in this process. RESULTS: Oleic acid inhibited platelet aggregation, granule release, and calcium mobilization. Furthermore, it inhibited the spread of platelets on fibrinogen. We also found that oleic acid delayed arterial thrombosis in mice, as demonstrated in a murine model of ferric chloride-induced carotid artery thrombosis. The molecular mechanism of its inhibition of platelet activity may be through the Syk-PLCγ2 and CaMKKß/AMPKα/VASP pathways. In addition, we demonstrated that the phosphorylation of AMPK at Ser496 was an important mechanism of platelet activation. CONCLUSIONS: Our study showed that oleic acid inhibits platelet activation and reduces thrombogenesis by inhibiting the phosphorylation of multiple signaling molecules, offering new insights into the research and development of antiplatelet drugs. Video Abstract.


Subject(s)
Oleic Acid , Thrombosis , Humans , Mice , Animals , Oleic Acid/pharmacology , Oleic Acid/metabolism , Platelet Activation , Blood Platelets , Thrombosis/metabolism , Phosphorylation , Collagen/metabolism , Fibrinogen/metabolism , Fibrinogen/pharmacology
6.
Neurochem Res ; 48(11): 3255-3269, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37442896

ABSTRACT

Multiple sclerosis (MS) is an autoimmune demyelinating neurodegenerative disease of the central nervous system (CNS) due to injury of the myelin sheath by immune cells. The clotting factor fibrinogen is involved in the pathogenesis of MS by triggering microglia and the progress of neuroinflammation. Fibrinogen level is correlated with MS severity; consequently, inhibition of the fibrinogen cascade may reduce MS neuropathology. Thus, this review aimed to clarify the potential role of fibrinogen in the pathogenesis of MS and how targeting of fibrinogen affects MS neuropathology. Accumulation of fibrinogen in the CNS may occur independently or due to disruption of blood-brain barrier (BBB) integrity in MS. Fibrinogen acts as transduction and increases microglia activation which induces the progression of inflammation, oxidative stress, and neuronal injury. Besides, brain fibrinogen impairs the remyelination process by inhibiting the differentiation of oligodendrocyte precursor cells. These findings proposed that fibrinogen is associated with MS neuropathology through interruption of BBB integrity, induction of neuroinflammation, and demyelination with inhibition of the remyelination process by suppressing oligodendrocytes. Therefore, targeting of fibrinogen and/or CD11b/CD18 receptors by metformin and statins might decrease MS neuropathology. In conclusion, inhibiting the expression of CD11b/CD18 receptors by metformin and statins may decrease the pro-inflammatory effect of fibrinogen on microglia which is involved in the progression of MS.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Multiple Sclerosis , Neurodegenerative Diseases , Humans , Multiple Sclerosis/metabolism , Fibrinogen/metabolism , Fibrinogen/pharmacology , Neuroinflammatory Diseases , Neurodegenerative Diseases/metabolism , Myelin Sheath/metabolism , Fear
7.
BMC Genomics ; 23(1): 578, 2022 Aug 11.
Article in English | MEDLINE | ID: mdl-35953779

ABSTRACT

Berberine hydrochloride is the main effective component of Coptis spp. used in Chinese herbal medicine and its underlying molecular mechanisms, responsible for inducing effects in crustacean species, are not fully understood. In this study, the molecular response of the crab Charybdis japonica to berberine hydrochloride exposure was studied using transcriptome sequencing. The survival rate, gene expression and activities of several immune enzymes were measured after berberine hydrochloride treatments, with or without injection of the pathogenic bacterium Aeromonas hydrophila. A total of 962 differentially expressed genes (464 up-regulated and 498 down-regulated) were observed during exposure to 100 mg/L of berberine hydrochloride and in the control group after 48 h. Enrichment analysis revealed that these genes are involved in metabolism, cellular processes, signal transduction and immune functions, indicating that exposure to berberine hydrochloride activated the immune complement system. This bioactive compound simultaneously activated fibrinogen beta (FGB), fibrinogen alpha (FGA), alpha-2-macroglobulin (A2M), kininogen (KNG), fibrinogen gamma chain (FGB), alpha-2-HS-glycoprotein (AHSG), caspase-8 (CASP8), cathepsin L (CTSL), adenylate cyclase 3 (Adcy3) and MMP1. Its action could significantly increase the survival rate of the crabs injected with A. hydrophila and promote the activity of LZM, Caspas8, FGA, ACP and AKP in the hepatopancreas. When A. hydrophila was added, the neutralization of 300 mg/L berberine hydrochloride maximized the activities of Caspas8, LZM, ACP and AKP. Our results provide a new understanding of the potential effects of berberine hydrochloride on the immune system mechanisms in crustaceans.


Subject(s)
Berberine , Brachyura , Animals , Berberine/pharmacology , Brachyura/genetics , Fibrinogen/pharmacology , Hepatopancreas , Immunity/genetics
8.
J Pharmacol Exp Ther ; 383(1): 2-10, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35963618

ABSTRACT

N-stearoylethanolamine (NSE), a lipid mediator that belongs to the N-acylethanolamine (NAE) family, has anti-inflammatory, antioxidant, and membranoprotective actions. In contrast to other NAEs, NSE does not interact with cannabinoid receptors. The exact mechanism of its action remains unclear. The aim of this study is to evaluate the action of NSE on activation, aggregation, and adhesion of platelets that were chosen as a model of cellular response. Aggregation of platelets was measured to analyze the action of NSE (10-6-10-10 M) on platelet reactivity. Changes in granularity and shape of resting platelets and platelets stimulated with ADP in the presence of NSE were monitored by flow cytometry, and platelet deganulation was monitored by spectrofluorimetry. In vivo studies were performed using obese insulin-resistant rats. Binding of fibrinogen to the GPIIb/IIIa receptor was estimated using indirect ELISA and a scanning electron microscopy (SEM). It was found that NSE inhibits the activation and aggregation of human platelets. Our results suggest that NSE may decrease the activation and subsequent aggregation of platelets induced by ristocetin, epinephrine, and low doses of ADP. NSE also reduced the binding of fibrinogen to GPIIb/IIIa on activated platelets. These effects could be explained by the inhibition of platelet activation mediated by integrin receptors: the GPIb-IX-V complex for ristocetin-induced activation and GPIIb/IIIa when epinephrine and low doses of ADP were applied. The anti-platelet effect of NSE complements its anti-inflammatory effect and allows us to prioritize studies of NSE as a potent anti-thrombotic agent. SIGNIFICANCE STATEMENT: N-stearoylethanolamine (NSE) was shown to possess inhibitory action on platelet activation, adhesion, and aggregation. The mechanism of inhibition possibly involves integrin receptors. This finding complements the known anti-inflammatory effects of NSE.


Subject(s)
Platelet Aggregation , Ristocetin , Adenosine Diphosphate/metabolism , Adenosine Diphosphate/pharmacology , Animals , Blood Platelets , Epinephrine/metabolism , Epinephrine/pharmacology , Ethanolamines , Fibrinogen/metabolism , Fibrinogen/pharmacology , Humans , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/pharmacology , Rats , Ristocetin/metabolism , Ristocetin/pharmacology , Stearic Acids
9.
Liver Int ; 42(2): 435-443, 2022 02.
Article in English | MEDLINE | ID: mdl-34894081

ABSTRACT

BACKGROUND & AIMS: Patients with liver disease may acquire substantial changes in their hemostatic system, which are most pronounced in patients who are critically ill. Changes in the quality of the fibrin clot in critically ill patients have not been studied in detail. Here we assessed markers of fibrin clot quality and effects of coagulation factor concentrates in patients with acutely decompensated (AD) cirrhosis and acute on chronic liver failure (ACLF). METHODS: We measured plasma levels of fibrinogen, factor XIII, prothrombin and performed thrombin generation assays in 52 AD patients, 58 ACLF patients and 40 controls. In addition, we examined the effects of coagulation factor concentrates on functional assays of fibrin quality. RESULTS: We found increased thrombin generating capacity in both AD and ACLF in comparison with healthy controls. Plasma levels of prothrombin, fibrinogen, and factor XIII were lower in patients compared to controls, appeared lower in ACLF compared to AD patients, and were related to clinical outcomes. Fibrinogen concentrate, but not factor XIII or prothrombin complex concentrate, improved clot quality in vitro. Prothrombin complex concentrate increased the resistance of the clot to break down. CONCLUSIONS: We have demonstrated elevated thrombin generation but decreased plasma levels of prothrombin, fibrinogen and FXIII in acutely ill patients with cirrhosis. In addition, we showed that fibrinogen concentrate and PCCs, but not factor XIII concentrate, improve clot properties in patient plasma. Whether there is true clinical benefit from coagulation factor concentrates in prevention or treatment of bleeding requires further study. LAY SUMMARY: Patients with liver diseases are at risk of bleeding, but mechanisms involved in this bleeding risk are incompletely understood. We studied components that determine the stability of the blood clot and found that concentrations of certain proteins involved in clot stability are present in low levels in acutely ill patients with liver disease. We furthermore demonstrated that some clinically available drugs improve the stability of blood clots from these patients in a test tube.


Subject(s)
Fibrin , Thrombosis , Blood Coagulation , Blood Coagulation Factors/therapeutic use , Fibrin/metabolism , Fibrinogen/metabolism , Fibrinogen/pharmacology , Humans , Liver Cirrhosis/complications , Liver Cirrhosis/drug therapy
10.
Int J Med Sci ; 19(1): 195-204, 2022.
Article in English | MEDLINE | ID: mdl-34975313

ABSTRACT

Human fibroleukin 2 (Fgl2), a member of the fibrinogen superfamily, can cleave prothrombin to generate thrombin or is secreted in a soluble form as a new type of effector of Tregs with immunomodulatory functions. However, there is little research on the role of Fgl2 in cutaneous squamous cell carcinoma (CSCC) growth. We examined the expression of Fgl2 in samples from CSCC patients and CSCC cell lines. Then, the effect of Fgl2 on CSCC was evaluated in vitro and in animals. Regulation of autophagy by Fgl2 was explored in CSCC. Coimmunoprecipitation (Co-IP) and immunofluorescence colocalization experiments were conducted to identify the regulatory effect of Fgl2 on the downstream protein Tyrobp. Then, gain- or loss-of-function analyses and evaluation of Tyrobp expression were performed to validate its role in autophagy and proliferation promoted by Fgl2. Here, our study demonstrated that Fgl2 promoted the proliferation of CSCC cells in vitro and in vivo. Knocking down Fgl2 reduced CSCC cell proliferation and inhibited autophagy in CSCC. Mechanistically, Fgl2 interacted with Tyrobp and promoted ERK-dependent autophagy, resulting in the proliferation of CSCC cells. Our study suggested that Fgl2 could be a promising prognostic biomarker and useful therapeutic target for CSCC.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carcinoma, Squamous Cell/pathology , Fibrinogen/metabolism , Membrane Proteins/metabolism , Skin Neoplasms/pathology , Animals , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Proliferation , Fibrinogen/pharmacology , Humans , MAP Kinase Signaling System , Signal Transduction , Skin Neoplasms/metabolism , Up-Regulation
11.
Transfusion ; 61(7): 2179-2194, 2021 07.
Article in English | MEDLINE | ID: mdl-33948950

ABSTRACT

BACKGROUND: Platelet transfusion is challenging in emergency medicine because of short platelet shelf life and stringent storage conditions. Platelet-derived extracellular vesicles (PEV) exhibit platelet-like properties. A plasma generated from expired platelet units rich in procoagulant PEV may be able to combine the benefits of plasma and platelets for resuscitation while increasing shelf life and utilizing an otherwise wasted resource. STUDY DESIGN AND METHODS: Freeze-thaw cycling of platelet-rich plasma (PRP) followed by centrifugation to remove platelet remnants was utilized to generate platelet-enhanced plasma (PEP). An in vitro model of dilutional coagulopathy was also designed and used to test PEP. Rotational thromboelastometry and calibrated automated thrombography were used to assess clotting and extracellular vesicles (EV) procoagulant activity. Capture arrays were used to specifically measure EV subpopulations of interest (ExoView™, NanoView Biosciences). Captured vesicles were quantified and labeled with Annexin-V-FITC, CD41-PE, and CD63-AF647. Platelet alpha granule content (platelet-derived growth factor AB, soluble P-selectin, vascular endothelial growth factor A, and neutrophil activating peptide 2-chemokine (C-X-C motif) ligand 7) was measured. Commercially available platelet lysates were also characterized. RESULTS: PEP is highly procoagulant, rich in growth factors, exhibits enhanced thrombin generation, and restores hemostasis within an in vitro model of dilutional coagulopathy. The predominant vesicle population were PEV with 7.0 × 109 CD41+PS+ EV/ml compared to 4.7 × 107 CD41+PS+ EV/ml in platelet-free plasma (p = .0079). Commercial lysates show impaired but rescuable clotting. DISCUSSION: PEP is a unique candidate resuscitation fluid containing high PEV concentration with preliminary evidence, indicating a potential for upscaling the approach using platelet concentrates. Commercial lysate manufacturer workflows may be suitable for this, but further optimization and characterization of PEP is required.


Subject(s)
Blood Coagulation , Extracellular Vesicles/transplantation , Plasma , Platelet Transfusion , Resuscitation , Thrombin/biosynthesis , Blood Cell Count , Blood Platelets , Blood Preservation/methods , Fibrinogen/analysis , Fibrinogen/pharmacology , Humans , Intercellular Signaling Peptides and Proteins/blood , P-Selectin/blood , Partial Thromboplastin Time , Platelet Membrane Glycoprotein IIb/blood , Platelet-Rich Plasma , Prothrombin Time , Temperature , Thrombelastography , Time Factors
12.
Transfusion ; 61(6): 1943-1954, 2021 06.
Article in English | MEDLINE | ID: mdl-33755208

ABSTRACT

BACKGROUND: Fibrinogen concentrates and cryoprecipitate are currently used for fibrinogen supplementation in bleeding patients with dysfibrinogenemia. Both products provide an abundant source of fibrinogen but take greater than 10 min to prepare for administration. Fibrinogen concentrates lack coagulation factors (i.e., factor VIII [FVIII], factor XIII [FXIII], von Willebrand factor [VWF]) important for robust hemostatic function. Cryoprecipitate products contain these factors but have short shelf lives (<6 h). Pathogen reduction (PR) of cryoprecipitate would provide a shelf-stable immediately available adjunct containing factors important for rescuing hemostatic dysfunction. STUDY DESIGN AND METHODS: Hemostatic adjunct study products were psoralen-treated PR-cryoprecipitated fibrinogen complex (PR-Cryo FC), cryoprecipitate (Cryo), and fibrinogen concentrates (FibCon). PR-Cryo FC and Cryo were stored for 10 days at 20-24°C. Adjuncts were added to coagulopathies (dilutional, 3:7 whole blood [WB]:normal saline; or lytic, WB + 75 ng/ml tissue plasminogen activator), and hemostatic function was assessed by rotational thromboelastometry and thrombin generation. RESULTS: PR of cryoprecipitate did not reduce levels of FVIII, FXIII, or VWF. PR-Cryo FC rescued dilutional coagulopathy similarly to Cryo, while generating significantly more thrombin than FibCon, which also rescued dilutional coagulopathy. Storage out to 10 days at 20-24°C did not diminish the hemostatic function of PR-Cryo FC. DISCUSSION: PR-Cryo FC provides similar and/or improved hemostatic rescue compared to FibCon in dilutional coagulopathies, and this rescue ability is stable over 10 days of storage. In hemorrhaging patients, where every minute delay is associated with a 5% increase in mortality, the immediate availability of PR-Cryo FC has the potential to improve outcomes.


Subject(s)
Blood Safety , Factor VIII/pharmacology , Fibrinogen/pharmacology , Hemostasis , Hemostatics/pharmacology , Blood Coagulation Disorders/blood , Blood Coagulation Disorders/therapy , Blood Coagulation Factors/analysis , Blood Coagulation Factors/pharmacology , Blood Safety/methods , Factor VIII/analysis , Fibrinogen/analysis , Hemostasis/drug effects , Hemostatics/analysis , Humans , Sterilization/methods
13.
Transfusion ; 61(2): 557-567, 2021 02.
Article in English | MEDLINE | ID: mdl-33247486

ABSTRACT

BACKGROUND: Cold storage of platelets (PLTs) has the potential advantage of prolonging storage time while reducing posttransfusion infection given the decreased likelihood of bacterial outgrowth during storage and possibly beneficial effects in treating bleeding patients. However, cold storage reduces PLT survival through the induction of complex storage lesions, which are more accentuated when storage is prolonged. STUDY DESIGN AND METHODS: Whole blood-derived PLT-rich plasma concentrates from seven PLT pools (n = 5 donors per pool). PLT additive solution was added (67%/33% plasma) and the product was split into 50-mL bags. Split units were stored in the presence or absence of 1 mM of N-acetylcysteine (NAC) under agitation for up to 14 days at room temperature or in the cold and were analyzed for PLT activation, fibrinogen-dependent spreading, microparticle formation, mitochondrial respiratory activity, reactive oxygen species (ROS) generation, as well as in vivo survival and bleeding time correction in immunodeficient mice. RESULTS: Cold storage of PLTs for 7 days or longer induces significant PLT activation, cytoskeletal damage, impaired fibrinogen spreading, enhances mitochondrial metabolic decoupling and ROS generation, and increases macrophage-dependent phagocytosis and macrophage-independent clearance. Addition of NAC prevents PLT clearance and allows a correction of the prolonged bleeding time in thrombocytopenic, aspirin-treated, immunodeficient mice. CONCLUSIONS: Long-term cold storage induces mitochondrial uncoupling and increased proton leak and ROS generation. The resulting ROS is a crucial contributor to the increased macrophage-dependent and -independent clearance of functional PLTs and can be prevented by the antioxidant NAC in a magnesium-containing additive solution.


Subject(s)
Acetylcysteine/pharmacology , Antioxidants/pharmacology , Blood Platelets/drug effects , Blood Preservation/methods , Mitochondria/metabolism , Animals , Aspirin/toxicity , Bleeding Time , Blood Platelets/ultrastructure , Cell Shape/drug effects , Cold Temperature , Fibrinogen/pharmacology , Humans , Macrophages/drug effects , Mice , Mice, Inbred NOD , Mice, SCID , Oxygen Consumption , Phagocytosis/drug effects , Platelet Activation/drug effects , Platelet Transfusion , Platelet-Rich Plasma , Reactive Oxygen Species/analysis , Thrombocytopenia/chemically induced , Thrombocytopenia/therapy
14.
Acta Anaesthesiol Scand ; 65(10): 1439-1446, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34368944

ABSTRACT

BACKGROUND: Fibrinogen concentrate is used clinically to improve hemostasis in bleeding patients. We investigated and compared the efficacy of three commercially available fibrinogen concentrates to improve clot strength in blood samples from cardiac surgery patients. OBJECTIVES: Postoperative blood samples were collected from 23 cardiac surgery patients. Samples were each divided into four vials, each supplemented with 1.125 mg of fibrinogen of one of three fibrinogen concentrates (RiaSTAP® , Fibryga® , FibCLOT® ), or placebo. The fibrinogen dose corresponded to 2.5 g per 70 kg of body weight. Clot strength after supplementation was assessed in duplicate with rotational thromboelastometry (ROTEM® ) using FIBTEM maximum clot firmness, EXTEM clot formation time, and maximum clot firmness assays. RESULTS: In vitro fibrinogen concentrate supplementation of the samples resulted in higher plasma fibrinogen concentrations and improved clot strength with all three concentrates. Supplementation with FibCLOT increased FIBTEM maximum clot firmness (+46% [25th-75th percentile 35-55] compared to placebo) significantly more than did supplementation with Fibryga (+26% [21-35]) and RiaSTAP (+29% [22-47], p < .001). FibCLOT supplementation also shortened EXTEM clot formation time and increased EXTEM maximum clot firmness to a greater extent than did the other concentrates (both p < .001). CONCLUSIONS: At the selected dose, FibCLOT was more effective than Fibryga and RiaSTAP in restoring clot strength in postoperative blood samples from cardiac surgery patients. These results may have implications for the choice of fibrinogen concentrate and dosing.


Subject(s)
Blood Coagulation , Cardiac Surgical Procedures , Fibrinogen/pharmacology , Hemostatics , Hemostatics/pharmacology , Humans , Thrombelastography
15.
Int J Mol Sci ; 22(24)2021 Dec 09.
Article in English | MEDLINE | ID: mdl-34948063

ABSTRACT

Traumatic injury of the oral cavity is atypical and often accompanied by uncontrolled bleeding and inflammation. Injectable hydrogels have been considered to be promising candidates for the treatment of oral injuries because of their simple formulation, minimally invasive application technique, and site-specific delivery. Fibrinogen-based hydrogels have been widely explored as effective materials for wound healing in tissue engineering due to their uniqueness. Recently, an injectable foam has taken the spotlight. However, the fibrin component of this biomaterial is relatively stiff. To address these challenges, we created keratin-conjugated fibrinogen (KRT-FIB). This study aimed to develop a novel keratin biomaterial and assess cell-biomaterial interactions. Consequently, a novel injectable KRT-FIB hydrogel was optimized through rheological measurements, and its injection performance, swelling behavior, and surface morphology were investigated. We observed an excellent cell viability, proliferation, and migration/cell-cell interaction, indicating that the novel KRT-FIB-injectable hydrogel is a promising platform for oral tissue regeneration with a high clinical applicability.


Subject(s)
Biocompatible Materials/pharmacology , Fibrinogen/pharmacology , Keratins, Hair-Specific/pharmacology , Wound Healing , Biocompatible Materials/chemistry , Cell Proliferation/drug effects , Cell Survival , Cells, Cultured , Fibrinogen/chemistry , Humans , Hydrogels , Injections , Keratins, Hair-Specific/chemistry , Porosity , Regeneration , Rheology , Viscosity
16.
Br J Haematol ; 189(2): 335-338, 2020 04.
Article in English | MEDLINE | ID: mdl-31792942

ABSTRACT

This study was conducted to evaluate the expression of fibrinogen receptors on platelets of Philadelphia-negative chronic myeloproliferative neoplasm (MPN) patients. We collected blood samples from 40 consecutive MPN patients and healthy volunteers. We performed flow cytometry analysis of P-selectin expression and integrin beta-3, activation of glycoprotein (GP) IIb/IIIa and fibrinogen receptor exposure (PAC-1 binding). Surprisingly, we found a very low PAC-1 binding capacity in MPN patients; however, the expression of PAC-1 was almost completely recovered with aspirin intake. We hypothesize that the hypercoagulable states observed in MPN patients could depend on a primarily plasma-driven impairment of fibrin turnover and thrombin generation.


Subject(s)
Aspirin/therapeutic use , Fibrinogen/therapeutic use , Myeloproliferative Disorders/drug therapy , Adult , Aged , Aged, 80 and over , Aspirin/pharmacology , Blood Platelets , Chronic Disease , Fibrinogen/pharmacology , Humans , Middle Aged
17.
Haemophilia ; 26(1): 25-32, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31709664

ABSTRACT

Hereditary fibrinogen disorders (HFD) are rare coagulation disorders. Even if the spectrum of symptoms is broad depending on the sub-type, bleeding is the most common complication. Of the available sources of fibrinogen replacement, fibrinogen concentrate provides a safer and more effective option to treat and prevent bleeding. Recent clinical trials on established and new fibrinogen concentrates have increased our knowledge on the clinical pharmacology of these products, pointing out possible age and weight differences for dose adjustment. The efficacy of fibrinogen infusions has been demonstrated, especially for the management of acute bleeding with an excellent response based on investigator rating. The target fibrinogen levels in the setting of both minor and major surgeries have been better specified. The safety has been confirmed with a low number of adverse events but there still remains concern over possible thrombotic risks. Pharmacological, clinical aspects and future perspectives on the utilization of fibrinogen concentrates in the treatment and prevention of bleeding in patients with HFD are reviewed.


Subject(s)
Blood Coagulation Disorders/congenital , Blood Coagulation Disorders/drug therapy , Fibrinogen/therapeutic use , Area Under Curve , Fibrinogen/adverse effects , Fibrinogen/pharmacokinetics , Fibrinogen/pharmacology , Humans , Treatment Outcome
18.
Nano Lett ; 19(9): 6554-6563, 2019 09 11.
Article in English | MEDLINE | ID: mdl-31418579

ABSTRACT

As a key player in blood coagulation and tissue repair, fibrinogen has gained increasing attention to develop nanofibrous biomaterial scaffolds for wound healing. Current techniques to prepare protein nanofibers, like electrospinning or extrusion, are known to induce lasting changes in the protein conformation. Often, such secondary changes are associated with amyloid transitions, which can evoke unwanted disease mechanisms. Starting from our recently introduced technique to self-assemble fibrinogen scaffolds in physiological salt buffers, we here investigated the morphology and secondary structure of our novel fibrinogen nanofibers. Aiming at optimum self-assembly conditions for wound healing scaffolds, we studied the influence of fibrinogen concentration and pH on the protein conformation. Using circular dichroism and Fourier-transform infrared spectroscopy, we observed partial transitions from α-helical structures to ß-strands upon fiber formation. Interestingly, a staining with thioflavin T revealed that this conformational transition was not associated with any amyloid formation. Toward novel scaffolds for wound healing, which are stable in aqueous environment, we also introduced cross-linking of fibrinogen scaffolds in formaldehyde vapor. This treatment allowed us to maintain the nanofibrous morphology while the conformation of fibrinogen nanofibers was redeveloped toward a more native state after rehydration. Altogether, self-assembled fibrinogen scaffolds are excellent candidates for novel wound healing systems since their multiscale structures can be well controlled without inducing any pathogenic amyloid transitions.


Subject(s)
Fibrinogen/chemistry , Nanofibers/chemistry , Wound Healing , Fibrinogen/pharmacology , Humans , Nanofibers/therapeutic use
19.
Cell Tissue Res ; 375(3): 709-721, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30338376

ABSTRACT

Full-thickness skin defect is one of the main clinical problems, which cannot be repaired spontaneously. The aim of this study was to evaluate the feasibility of combining nanofibers with ADM as a bilayer scaffold for treatment of full-thickness skin wounds in a single-step procedure. The nanofibrous polycaprolactone/fibrinogen scaffolds were fabricated by electrospinning. Subsequently, mesenchymal stem cells were isolated from rat adipose tissues and characterized by flow cytometry. Cell adhesion, proliferation, and the epidermal differentiation potential of adipose-derived stem cells (ADSCs) on nanofibrous scaffolds were investigated by scanning electron microscopy (SEM), alamarBlue, and real-time PCR, respectively. In animal studies, full-thickness excisional wounds were created on the back of rats and treated with following groups: ADM, ADM-ADSCs, nanofiber, nanofiber-ADSCs, bilayer, and bilayer-ADSCs. In all groups, wounds were harvested on days 14 and 21 after treatment to evaluate re-epithelialization, blood vessel density, and collagen content. The results indicated that ADSCs seeded on ADM, nanofiber, and bilayer scaffolds can promote re-epithelialization, angiogenesis, and collagen remodeling in comparison with cell-free scaffolds. In conclusion, nanofiber-ADSCs showed the best results for re-epithelialization (according to histological scoring), average blood vessel density (92.7 ± 6.8), and collagen density (87.4 ± 4.9%) when compared to the control and other experimental groups.


Subject(s)
Acellular Dermis/metabolism , Mesenchymal Stem Cells/cytology , Nanofibers/chemistry , Skin/pathology , Tissue Scaffolds/chemistry , Wound Healing , Acellular Dermis/drug effects , Adipose Tissue/cytology , Animals , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Collagen/metabolism , Epidermis/drug effects , Epidermis/metabolism , Fibrinogen/pharmacology , Mesenchymal Stem Cells/drug effects , Neovascularization, Physiologic/drug effects , Polyesters/pharmacology , Rats, Wistar , Tissue Engineering , Wound Healing/drug effects
20.
Transfusion ; 59(10): 3186-3196, 2019 10.
Article in English | MEDLINE | ID: mdl-31257633

ABSTRACT

BACKGROUND: We previously developed substitutes for red blood cells (RBCs) and platelets (PLTs) for transfusion. These substitutes included hemoglobin vesicles (HbVs) and fibrinogen γ-chain (dodecapeptide HHLGGAKQAGDV, H12)-coated, adenosine diphosphate (ADP)-encapsulated liposomes [H12-(ADP)-liposomes]. Here, we examined the efficacy of combination therapy using these substitutes instead of RBC and PLT transfusion in a rabbit model with trauma-induced massive hemorrhage with coagulopathy. STUDY DESIGN AND METHODS: Thrombocytopenia (PLT count approximately 40,000/µL) was induced in rabbits by repeated blood withdrawal and isovolemic transfusion with autologous RBCs. Thereafter, lethal hemorrhage was induced in rabbits by noncompressible penetrating liver injury. Subsequently, H12-(ADP)-liposomes with platelet-poor plasma (PPP), platelet-rich plasma (PRP), or PPP alone were administered to stop bleeding. Once achieving hemostasis, HbVs, allogenic RBCs, or 5% albumin were transfused into rabbits to rescue them from fatal anemia following massive hemorrhage. RESULTS: Administration of H12-(ADP)-liposomes/PPP as well as PRP (but not PPP) effectively stopped liver bleeding (100% hemostasis). The subsequent administration with HbVs as well as RBCs after hemostasis markedly rescued rabbits from fatal anemia (75% and 70% survivals for 24 hr, respectively). In contrast, 5% albumin administration rescued none of the rabbits. CONCLUSION: Combination therapy with H12-(ADP)-liposomes and HbVs may be effective for damage control resuscitation of trauma-induced massive hemorrhage.


Subject(s)
Adenosine Diphosphate/pharmacology , Blood Substitutes/pharmacology , Fibrinogen/pharmacology , Hemoglobins/pharmacology , Hemorrhage/drug therapy , Oligopeptides/pharmacology , Wounds and Injuries/drug therapy , Animals , Disease Models, Animal , Drug Therapy, Combination , Hemorrhage/blood , Hemorrhage/pathology , Liposomes , Rabbits , Wounds and Injuries/blood , Wounds and Injuries/pathology
SELECTION OF CITATIONS
SEARCH DETAIL