Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
3.
Brain Behav Immun ; 93: 288-298, 2021 03.
Article in English | MEDLINE | ID: mdl-33401017

ABSTRACT

Recently it was shown that the hematophagous salivary gland protein agaphelin exhibits multiple antithrombotic effects without promoting the risk of bleeding. Agaphelin inhibits neutrophil elastase and thereby reduces cathepsin G-induced platelet aggregation. However, it is still unclear, whether pharmacological treatment with agaphelin in brain ischemia is protective and, regarding its bleeding risk, safe. To elucidate this issue, male C57BL/6 mice were subjected to 60 min of transient middle cerebral artery occlusion (tMCAO) and treated with 0.25 mg/kg agaphelin intravenously immediately after tMCAO. On day 1 and 7, infarct volume and functional neurological outcome were assessed by behavioural tests, histochemistry and magnetic resonance imaging. Thrombus formation, intracerebral bleeding risk, blood-brain barrier damage and the local inflammatory response were determined on day 1. This study shows for the first time a protective effect of agaphelin characterized by smaller infarct volume, reduced neurological deficits and reduced animal mortality. This protective effect was associated with reduced local thrombus formation, increased blood-brain barrier integrity and reduced brain inflammatory response. It is essential to mention that the protective effect of agaphelin was not linked to an increased risk of intracerebral bleeding. The promotion of brain tissue survival and inhibition of thromboinflammation identifies agaphelin as a promising treatment option in ischemic stroke, which considering the lack of bleeding risk should potentially be safe.


Subject(s)
Brain Ischemia , Insect Proteins/pharmacology , Ischemic Stroke , Pancreatic Elastase/antagonists & inhibitors , Salivary Proteins and Peptides/pharmacology , Thrombosis , Animals , Blood-Brain Barrier , Disease Models, Animal , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/drug therapy , Inflammation/drug therapy , Ischemic Stroke/drug therapy , Male , Mice , Mice, Inbred C57BL
4.
Front Cardiovasc Med ; 8: 785738, 2021.
Article in English | MEDLINE | ID: mdl-34977191

ABSTRACT

The novel coronavirus disease (COVID-19) is associated with a high incidence of coagulopathy and venous thromboembolism that may contribute to the worsening of the clinical outcome in affected patients. Marked increased D-dimer levels are the most common laboratory finding and have been repeatedly reported in critically ill COVID-19 patients. The infection caused by Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) is followed by a massive release of pro-inflammatory cytokines, which mediate the activation of endothelial cells, platelets, monocytes, and neutrophils in the vasculature. In this context, COVID-19-associated thrombosis is a complex process that seems to engage vascular cells along with soluble plasma factors, including the coagulation cascade, and complement system that contribute to the establishment of the prothrombotic state. In this review, we summarize the main findings concerning the cellular mechanisms proposed for the establishment of COVID-19-associated thrombosis.

5.
Front Immunol ; 10: 2088, 2019.
Article in English | MEDLINE | ID: mdl-31552036

ABSTRACT

Cancer patients are at increased risk of developing thrombosis, comorbidity that has been associated with increased neutrophil counts and the formation of neutrophil extracellular traps (NETs). Interleukin-1ß (IL-1ß) modulates the expression of granulocyte colony-stimulating factor (G-CSF), a cytokine that promotes cancer-associated neutrophilia and NET generation. Herein, we combined a murine breast cancer model with a flow-restriction thrombosis model to evaluate whether the IL-1ß blockade could interfere with cancer-associated thrombosis. Mice bearing metastatic 4T1 tumors exhibited high neutrophil counts as well as elevated expression of G-CSF and IL-1ß in their tumors. On the other hand, mice bearing non-metastatic 67NR tumors showed no elevation in neutrophil counts and displayed low expression levels of G-CSF and IL-1ß in their tumors. 4T1 tumor-bearing mice but not 67NR tumor-bearing mice exhibited a NET-dependent prothrombotic state. Pharmacological blockade of IL-1 receptor (IL-1R) decreased the primary growth of 4T1 tumors and reduced the systemic levels of myeloperoxidase, cell-free DNA (cfDNA) and G-CSF, without interfering with the neutrophil counts. Most remarkably, the blockade of IL-1R abolished the prothrombotic state observed in 4T1 tumor-bearing mice. Overall, our results demonstrate that IL-1ß might be a feasible target to attenuate cancer-associated thrombosis, particularly in cancer types that rely on increased G-CSF production and involvement of NET formation.


Subject(s)
Extracellular Traps/drug effects , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin-1beta/antagonists & inhibitors , Mammary Neoplasms, Experimental/complications , Receptors, Interleukin-1/antagonists & inhibitors , Thrombosis/prevention & control , Animals , Breast Neoplasms/complications , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Extracellular Traps/metabolism , Female , Granulocyte Colony-Stimulating Factor/genetics , Granulocyte Colony-Stimulating Factor/metabolism , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Leukocyte Count , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice, Inbred BALB C , Neutrophils/metabolism , Peroxidase/metabolism , Receptors, Interleukin-1/metabolism , Thrombosis/complications , Thrombosis/metabolism , Tumor Burden/drug effects
6.
Circ Res ; 125(5): 507-519, 2019 08 16.
Article in English | MEDLINE | ID: mdl-31248335

ABSTRACT

RATIONALE: PAD4 (peptidylarginine deiminase type IV), an enzyme essential for neutrophil extracellular trap formation (NETosis), is released together with neutrophil extracellular traps into the extracellular milieu. It citrullinates histones and holds the potential to citrullinate other protein targets. While NETosis is implicated in thrombosis, the impact of the released PAD4 is unknown. OBJECTIVE: This study tests the hypothesis that extracellular PAD4, released during inflammatory responses, citrullinates plasma proteins, thus affecting thrombus formation. METHODS AND RESULTS: Here, we show that injection of r-huPAD4 in vivo induces the formation of VWF (von Willebrand factor)-platelet strings in mesenteric venules and that this is dependent on PAD4 enzymatic activity. VWF-platelet strings are naturally cleaved by ADAMTS13 (a disintegrin and metalloproteinase with thrombospondin type-1 motif-13). We detected a reduction of endogenous ADAMTS13 activity in the plasma of wild-type mice injected with r-huPAD4. Using mass spectrometry and in vitro studies, we found that r-huPAD4 citrullinates ADAMTS13 on specific arginine residues and that this modification dramatically inhibits ADAMTS13 enzymatic activity. Elevated citrullination of ADAMTS13 was observed in plasma samples of patients with sepsis or noninfected patients who were elderly (eg, age >65 years) and had underlying comorbidities (eg, diabetes mellitus and hypertension) as compared with healthy donors. This shows that ADAMTS13 is citrullinated in vivo. VWF-platelet strings that form on venules of Adamts13-/- mice were immediately cleared after injection of r-huADAMTS13, while they persisted in vessels of mice injected with citrullinated r-huADAMTS13. Next, we assessed the effect of extracellular PAD4 on platelet-plug formation after ferric chloride-induced injury of mesenteric venules. Administration of r-huPAD4 decreased time to vessel occlusion and significantly reduced thrombus embolization. CONCLUSIONS: Our data indicate that PAD4 in circulation reduces VWF-platelet string clearance and accelerates the formation of a stable platelet plug after vessel injury. We propose that this effect is, at least in part, due to ADAMTS13 inhibition.


Subject(s)
Blood Platelets/metabolism , Protein-Arginine Deiminase Type 4/blood , Thrombosis/blood , Vascular System Injuries/blood , von Willebrand Factor/metabolism , Aged , Animals , Blood Platelets/drug effects , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein-Arginine Deiminase Type 4/toxicity , Thrombosis/chemically induced , Vascular System Injuries/chemically induced , Young Adult
7.
Sci Rep ; 8(1): 4806, 2018 03 19.
Article in English | MEDLINE | ID: mdl-29555911

ABSTRACT

Tick saliva is a rich source of modulators of vascular biology. We have characterized Ixonnexin, a member of the "Basic-tail" family of salivary proteins from the tick Ixodes scapularis. Ixonnexin is a 104 residues (11.8 KDa), non-enzymatic basic protein which contains 3 disulfide bonds and a C-terminal rich in lysine. It is homologous to SALP14, a tick salivary FXa anticoagulant. Ixonnexin was produced by ligation of synthesized fragments (51-104) and (1-50) followed by folding. Ixonnexin, like SALP14, interacts with FXa. Notably, Ixonnexin also modulates fibrinolysis in vitro by a unique salivary mechanism. Accordingly, it accelerates plasminogen activation by tissue-type plasminogen activator (t-PA) with Km 100 nM; however, it does not affect urokinase-mediated fibrinolysis. Additionally, lysine analogue ε-aminocaproic acid inhibits Ixonnexin-mediated plasmin generation implying that lysine-binding sites of Kringle domain(s) of plasminogen or t-PA are involved in this process. Moreover, surface plasmon resonance experiments shows that Ixonnexin binds t-PA, and plasminogen (KD 10 nM), but not urokinase. These results imply that Ixonnexin promotes fibrinolysis by supporting the interaction of plasminogen with t-PA through formation of an enzymatically productive ternary complex. Finally, in vivo experiments demonstrates that Ixonnexin inhibits FeCl3-induced thrombosis in mice. Ixonnexin emerges as novel modulator of fibrinolysis which may also affect parasite-vector-host interactions.


Subject(s)
Arterial Occlusive Diseases/prevention & control , Fibrinolysis/drug effects , Plasminogen/metabolism , Saliva/metabolism , Salivary Proteins and Peptides/pharmacology , Thrombosis/prevention & control , Ticks/metabolism , Tissue Plasminogen Activator/metabolism , Animals , Arterial Occlusive Diseases/chemically induced , Arterial Occlusive Diseases/pathology , Chlorides/toxicity , Ferric Compounds/toxicity , Mice , Noxae/toxicity , Thrombosis/chemically induced , Thrombosis/pathology
8.
Sci Rep ; 7(1): 6438, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28743887

ABSTRACT

Cancer patients are at an increased risk of developing thromboembolic complications. Several mechanisms have been proposed to explain cancer-associated thrombosis including the release of tumor-derived extracellular vesicles and the activation of host vascular cells. It was proposed that neutrophil extracellular traps (NETs) contribute to the prothrombotic phenotype in cancer. In this study, we evaluated the possible cooperation between tumor-derived exosomes and NETs in cancer-associated thrombosis. Female BALB/c mice were orthotopically injected with 4T1 breast cancer cells. The tumor-bearing animals exhibited increased levels of plasma DNA and myeloperoxidase in addition to significantly increased numbers of circulating neutrophils. Mice were subjected to either Rose Bengal/laser-induced venous thrombosis or ferric chloride-induced arterial thrombosis models. The tumor-bearing mice exhibited accelerated thrombus formation in both models compared to tumor-free animals. Treatment with recombinant human DNase 1 reversed the prothrombotic phenotype of tumor-bearing mice in both models. Remarkably, 4T1-derived exosomes induced NET formation in neutrophils from mice treated with granulocyte colony-stimulating factor (G-CSF). In addition, tumor-derived exosomes interacted with NETs under static conditions. Accordingly, the intravenous administration of 4T1-derived exosomes into G-CSF-treated mice significantly accelerated venous thrombosis in vivo. Taken together, our observations suggest that tumor-derived exosomes and neutrophils may act cooperatively in the establishment of cancer-associated thrombosis.


Subject(s)
Exosomes/pathology , Mammary Neoplasms, Experimental/pathology , Neutrophils/pathology , Thrombosis/etiology , Animals , Cell Line, Tumor , Disease Models, Animal , Extracellular Traps , Female , Granulocyte Colony-Stimulating Factor/pharmacology , Mammary Neoplasms, Experimental/complications , Mice, Inbred BALB C , Thrombosis/drug therapy , Venous Thrombosis/drug therapy , Venous Thrombosis/etiology
9.
Nucleic Acids Res ; 44(17): 8052-64, 2016 09 30.
Article in English | MEDLINE | ID: mdl-27566147

ABSTRACT

RNA aptamers are synthetic oligonucleotide-based affinity molecules that utilize unique three-dimensional structures for their affinity and specificity to a target such as a protein. They hold the promise of numerous advantages over biologically produced antibodies; however, the binding affinity and specificity of RNA aptamers are often insufficient for successful implementation in diagnostic assays or as therapeutic agents. Strong binding affinity is important to improve the downstream applications. We report here the use of the phosphorodithioate (PS2) substitution on a single nucleotide of RNA aptamers to dramatically improve target binding affinity by ∼1000-fold (from nanomolar to picomolar). An X-ray co-crystal structure of the α-thrombin:PS2-aptamer complex reveals a localized induced-fit rearrangement of the PS2-containing nucleotide which leads to enhanced target interaction. High-level quantum mechanical calculations for model systems that mimic the PS2 moiety and phenylalanine demonstrate that an edge-on interaction between sulfur and the aromatic ring is quite favorable, and also confirm that the sulfur analogs are much more polarizable than the corresponding phosphates. This favorable interaction involving the sulfur atom is likely even more significant in the full aptamer-protein complexes than in the model systems.


Subject(s)
Phosphates/metabolism , RNA/metabolism , Aptamers, Nucleotide , Cell Line , Humans , Kinetics , Limit of Detection , Models, Molecular , Nucleic Acid Conformation , Protein Binding , Proteins/metabolism , RNA Stability , Reference Standards , Serum/metabolism , Thermodynamics , Vascular Endothelial Growth Factor A/metabolism
10.
PLoS Negl Trop Dis ; 10(1): e0004298, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26745503

ABSTRACT

BACKGROUND: Hematophagous mosquitos and ticks avoid host hemostatic system through expression of enzyme inhibitors targeting proteolytic reactions of the coagulation and complement cascades. While most inhibitors characterized to date were found in the salivary glands, relatively few others have been identified in the midgut. Among those, Boophilin is a 2-Kunitz multifunctional inhibitor targeting thrombin, elastase, and kallikrein. However, the kinetics of Boophilin interaction with these enzymes, how it modulates platelet function, and whether it inhibits thrombosis in vivo have not been determined. METHODOLOGY/PRINCIPAL FINDINGS: Boophilin was expressed in HEK293 cells and purified to homogeneity. Using amidolytic assays and surface plasmon resonance experiments, we have demonstrated that Boophilin behaves as a classical, non-competitive inhibitor of thrombin with respect to small chromogenic substrates by a mechanism dependent on both exosite-1 and catalytic site. Inhibition is accompanied by blockade of platelet aggregation, fibrin formation, and clot-bound thrombin in vitro. Notably, we also identified Boophilin as a non-competitive inhibitor of FXIa, preventing FIX activation. In addition, Boophilin inhibits kallikrein activity and the reciprocal activation, indicating that it targets the contact pathway. Furthermore, Boophilin abrogates cathepsin G- and plasmin-induced platelet aggregation and partially affects elastase-mediated cleavage of Tissue Factor Pathway Inhibitor (TFPI). Finally, Boophilin inhibits carotid artery occlusion in vivo triggered by FeCl3, and promotes bleeding according to the mice tail transection method. CONCLUSION/SIGNIFICANCE: Through inhibition of several enzymes involved in proteolytic cascades and cell activation, Boophilin plays a major role in keeping the midgut microenvironment at low hemostatic and inflammatory tonus. This response allows ticks to successfully digest a blood meal which is critical for metabolism and egg development. Boophilin is the first tick midgut FXIa anticoagulant also found to inhibit thrombosis.


Subject(s)
Fibrinolytic Agents/isolation & purification , Fibrinolytic Agents/metabolism , Protease Inhibitors/isolation & purification , Protease Inhibitors/metabolism , Rhipicephalus/chemistry , Animals , Cell Line , Factor XIa/antagonists & inhibitors , Gastrointestinal Tract/chemistry , Gene Expression , Humans , Kallikreins/antagonists & inhibitors , Mice , Platelet Aggregation/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Thrombin/antagonists & inhibitors , Thrombosis/chemically induced , Thrombosis/prevention & control
11.
PLoS One ; 10(8): e0133991, 2015.
Article in English | MEDLINE | ID: mdl-26244557

ABSTRACT

A group of peptides from the salivary gland of the tick Hyalomma marginatum rufipes, a vector of Crimean Congo hemorrhagic fever show weak similarity to the madanins, a group of thrombin-inhibitory peptides from a second tick species, Haemaphysalis longicornis. We have evaluated the anti-serine protease activity of one of these H. marginatum peptides that has been given the name hyalomin-1. Hyalomin-1 was found to be a selective inhibitor of thrombin, blocking coagulation of plasma and inhibiting S2238 hydrolysis in a competitive manner with an inhibition constant (Ki) of 12 nM at an ionic strength of 150 mM. It also blocks the thrombin-mediated activation of coagulation factor XI, thrombin-mediated platelet aggregation, and the activation of coagulation factor V by thrombin. Hyalomin-1 is cleaved at a canonical thrombin cleavage site but the cleaved products do not inhibit coagulation. However, the C-terminal cleavage product showed non-competitive inhibition of S2238 hydrolysis. A peptide combining the N-terminal parts of the molecule with the cleavage region did not interact strongly with thrombin, but a 24-residue fragment containing the cleavage region and the C-terminal fragment inhibited the enzyme in a competitive manner and also inhibited coagulation of plasma. These results suggest that the peptide acts by binding to the active site as well as exosite I or the autolysis loop of thrombin. Injection of 2.5 mg/kg of hyalomin-1 increased arterial occlusion time in a mouse model of thrombosis, suggesting this peptide could be a candidate for clinical use as an antithrombotic.


Subject(s)
Anticoagulants/isolation & purification , Anticoagulants/pharmacology , Blood Coagulation/drug effects , Peptides/isolation & purification , Peptides/pharmacology , Thrombin/antagonists & inhibitors , Ticks/chemistry , Amino Acid Sequence , Animals , Anticoagulants/chemistry , Blood Coagulation Tests , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Peptides/chemistry , Platelet Aggregation/drug effects , Sequence Alignment , Thrombin/metabolism , Thrombosis/drug therapy
12.
PLoS Negl Trop Dis ; 9(6): e0003869, 2015.
Article in English | MEDLINE | ID: mdl-26110417

ABSTRACT

BACKGROUND: The saliva of blood-feeding arthropods contains a notable diversity of molecules that target the hemostatic and immune systems of the host. Dipetalodipin and triplatin are triatomine salivary proteins that exhibit high affinity binding to prostanoids, such as TXA2, thus resulting in potent inhibitory effect on platelet aggregation in vitro. It was recently demonstrated that platelet-derived TXA2 mediates the formation of neutrophil extracellular traps (NETs), a newly recognized link between inflammation and thrombosis that promote thrombus growth and stability. METHODOLOGY/PRINCIPAL FINDINGS: This study evaluated the ability of dipetalodipin and triplatin to block NETs formation in vitro. We also investigated the in vivo antithrombotic activity of TXA2 binding proteins by employing two murine models of experimental thrombosis. Remarkably, we observed that both inhibitors abolished the platelet-mediated formation of NETs in vitro. Dipetalodipin and triplatin significantly increased carotid artery occlusion time in a FeCl3-induced injury model. Treatment with TXA2-binding proteins also protected mice from lethal pulmonary thromboembolism evoked by the intravenous injection of collagen and epinephrine. Effective antithrombotic doses of dipetalodipin and triplatin did not increase blood loss, which was estimated using the tail transection method. CONCLUSIONS/SIGNIFICANCE: Salivary TXA2-binding proteins, dipetalodipin and triplatin, are capable to prevent platelet-mediated NETs formation in vitro. This ability may contribute to the antithrombotic effects in vivo. Notably, both molecules inhibit arterial thrombosis without promoting excessive bleeding. Our results provide new insight into the antihemostatic effects of TXA2-binding proteins and may have important significance in elucidating the mechanisms of saliva to avoid host's hemostatic responses and innate immune system.


Subject(s)
Insect Vectors/metabolism , Platelet Aggregation Inhibitors/pharmacology , Salivary Proteins and Peptides/pharmacology , Thrombosis/prevention & control , Thromboxane A2/metabolism , Triatominae/metabolism , Animals , Arthropod Proteins/metabolism , Arthropod Proteins/pharmacology , Blood Platelets/drug effects , Chagas Disease/transmission , Extracellular Traps/drug effects , Extracellular Traps/physiology , Female , Humans , Male , Mice , Mice, Inbred BALB C , Neutrophils/drug effects , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/metabolism , Protein Binding/drug effects , Recombinant Proteins , Salivary Proteins and Peptides/metabolism
13.
Thromb Res ; 136(2): 432-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26070446

ABSTRACT

BACKGROUND: The clotting initiator protein tissue factor (TF) has recently been described as a potential target that can be exploited to image aggressive tumors. Ixolaris is a specific TF inhibitor that blocks tumor cell procoagulant activity and tumor growth. OBJECTIVE: Herein we evaluated the ability of (99m)Tc-ixolaris to target tumor-derived TF using an orthotopic glioblastoma (GBM) model in mice. METHODS: The right forebrains of Swiss mice were stereotactically inoculated with U87-MG human GBM cells. Histological and immunohistochemical analyses were performed on the resulting tumors after 35-45 days. The biodistribution of (99m)Tc-ixolaris was evaluated by semi-quantitative whole-body scintigraphy and a quantitative analysis of radioactivity in isolated organs. RESULTS: No (99m)Tc-ixolaris uptake was observed in brain of tumor-free mice, independently of the integrity of brain-blood barrier. In contrast, the presence of TF-expressing brain tumor masses determined a significant (99m)Tc-ixolaris uptake. CONCLUSION: (99m)Tc-ixolaris recognized TF-expressing GBM cells in vivo. Given the proposed role of TF in tumor progression, (99m)Tc-ixolaris is a promising radiopharmaceutical agent for quantifying cancer-associated TF in aggressive tumors, including GBM.


Subject(s)
Biomarkers, Tumor/metabolism , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Salivary Proteins and Peptides/pharmacokinetics , Technetium/pharmacokinetics , Thromboplastin/metabolism , Animals , Brain Neoplasms/diagnostic imaging , Cell Line, Tumor , Glioblastoma/diagnostic imaging , Humans , Male , Metabolic Clearance Rate , Mice , Molecular Imaging/methods , Organ Specificity , Positron-Emission Tomography/methods , Radiopharmaceuticals/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity , Tissue Distribution
14.
PLoS Pathog ; 10(9): e1004338, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25211214

ABSTRACT

BACKGROUND: Invasion of mosquito salivary glands (SGs) by Plasmodium falciparum sporozoites is an essential step in the malaria life cycle. How infection modulates gene expression, and affects hematophagy remains unclear. PRINCIPAL FINDINGS: Using Affimetrix chip microarray, we found that at least 43 genes are differentially expressed in the glands of Plasmodium falciparum-infected Anopheles gambiae mosquitoes. Among the upregulated genes, one codes for Agaphelin, a 58-amino acid protein containing a single Kazal domain with a Leu in the P1 position. Agaphelin displays high homology to orthologs present in Aedes sp and Culex sp salivary glands, indicating an evolutionarily expanded family. Kinetics and surface plasmon resonance experiments determined that chemically synthesized Agaphelin behaves as a slow and tight inhibitor of neutrophil elastase (K(D) ∼ 10 nM), but does not affect other enzymes, nor promotes vasodilation, or exhibit antimicrobial activity. TAXIscan chamber assay revealed that Agaphelin inhibits neutrophil chemotaxis toward fMLP, affecting several parameter associated with cell migration. In addition, Agaphelin reduces paw edema formation and accumulation of tissue myeloperoxidase triggered by injection of carrageenan in mice. Agaphelin also blocks elastase/cathepsin-mediated platelet aggregation, abrogates elastase-mediated cleavage of tissue factor pathway inhibitor, and attenuates neutrophil-induced coagulation. Notably, Agaphelin inhibits neutrophil extracellular traps (NETs) formation and prevents FeCl3-induced arterial thrombosis, without impairing hemostasis. CONCLUSIONS: Blockade of neutrophil elastase emerges as a novel antihemostatic mechanism in hematophagy; it also supports the notion that neutrophils and the innate immune response are targets for antithrombotic therapy. In addition, Agaphelin is the first antihemostatic whose expression is induced by Plasmodium sp infection. These results suggest that an important interplay takes place in parasite-vector-host interactions.


Subject(s)
Anopheles/parasitology , Hemostasis/physiology , Host-Parasite Interactions , Insect Proteins/metabolism , Neutrophils/immunology , Plasmodium falciparum/pathogenicity , Salivary Proteins and Peptides/metabolism , Thrombosis/prevention & control , Amino Acid Sequence , Animals , Anopheles/metabolism , Circular Dichroism , Edema/etiology , Edema/metabolism , Edema/prevention & control , Female , Insect Proteins/chemistry , Insect Proteins/genetics , Insect Vectors , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Salivary Glands/metabolism , Salivary Glands/parasitology , Salivary Proteins and Peptides/chemistry , Salivary Proteins and Peptides/genetics , Sequence Homology, Amino Acid , Surface Plasmon Resonance
15.
Blood ; 122(25): 4094-106, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24159172

ABSTRACT

The identity of vampire bat saliva anticoagulant remained elusive for almost a century. Sequencing the salivary gland genes from the vampire bat Desmodus rotundus identified Desmolaris as a novel 21.5-kDa naturally deleted (Kunitz 1-domainless) form of tissue factor pathway inhibitor. Recombinant Desmolaris was expressed in HEK293 cells and characterized as a slow, tight, and noncompetitive inhibitor of factor (F) XIa by a mechanism modulated by heparin. Desmolaris also inhibits FXa with lower affinity, independently of protein S. In addition, Desmolaris binds kallikrein and reduces bradykinin generation in plasma activated with kaolin. Truncated and mutated forms of Desmolaris determined that Arg32 in the Kunitz-1 domain is critical for protease inhibition. Moreover, Kunitz-2 and the carboxyl-terminus domains mediate interaction of Desmolaris with heparin and are required for optimal inhibition of FXIa and FXa. Notably, Desmolaris (100 µg/kg) inhibited FeCl3-induced carotid artery thrombus without impairing hemostasis. These results imply that FXIa is the primary in vivo target for Desmolaris at antithrombotic concentrations. Desmolaris also reduces the polyphosphate-induced increase in vascular permeability and collagen- and epinephrine-mediated thromboembolism in mice. Desmolaris emerges as a novel anticoagulant targeting FXIa under conditions in which the coagulation activation, particularly the contact pathway, plays a major pathological role.


Subject(s)
Anticoagulants/chemistry , Anticoagulants/pharmacology , Chiroptera , Factor Xa Inhibitors , Salivary Proteins and Peptides/chemistry , Salivary Proteins and Peptides/pharmacology , Thrombosis/drug therapy , Animals , Bradykinin/chemistry , Bradykinin/genetics , Bradykinin/metabolism , Chlorides/adverse effects , Chlorides/pharmacology , Disease Models, Animal , Factor Xa/chemistry , Factor Xa/genetics , Factor Xa/metabolism , Ferric Compounds/adverse effects , Ferric Compounds/pharmacology , HEK293 Cells , Humans , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Kallikreins/chemistry , Kallikreins/genetics , Kallikreins/metabolism , Mice , Noxae/adverse effects , Noxae/pharmacology , Protein Structure, Tertiary , Salivary Proteins and Peptides/genetics , Thrombosis/chemically induced , Thrombosis/genetics , Thrombosis/metabolism
16.
Biochem Biophys Res Commun ; 436(2): 235-9, 2013 Jun 28.
Article in English | MEDLINE | ID: mdl-23726920

ABSTRACT

Aegyptin is a mosquito salivary gland protein and potent inhibitor of platelet aggregation. Aegyptin binds to the von Willebrand factor-binding site on collagen and prevents its interaction with platelets. Because collagen also induces plasma clotting by activation of factor XII, we evaluated the effects of aegyptin on collagen-induced coagulation activation and how it interferes with thrombosis in three different in vivo models. Our results demonstrate that aegyptin abolishes collagen-induced clot formation and thrombin generation in platelet-free plasma. Aegyptin has no antithrombotic activity in the arteriovenous shunt model (collagen-independent) but it prevents laser-induced collagen-mediated thrombus formation in rats. Furthermore, aegyptin protects mice from collagen and epinephrine-induced thromboembolism. Therefore, aegyptin has a dual antithrombotic mechanism: inhibition of platelet-collagen interaction and collagen's pro-coagulant activity. This is the first description of a collagen-binding protein that also inhibits collagen-mediated coagulant activity.


Subject(s)
Blood Coagulation/drug effects , Collagen/pharmacology , Insect Proteins/pharmacology , Pulmonary Embolism/prevention & control , Salivary Proteins and Peptides/pharmacology , Animals , Culicidae/metabolism , Dose-Response Relationship, Drug , Epinephrine/pharmacology , Female , HEK293 Cells , Humans , Insect Proteins/genetics , Male , Mice , Mice, Inbred BALB C , Rats , Rats, Wistar , Recombinant Proteins/pharmacology , Salivary Proteins and Peptides/genetics , Thrombin/metabolism
17.
An Acad Bras Cienc ; 85(1): 349-54, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23460444

ABSTRACT

Amylin is a 37-aminoacid pancreatic protein that exerts control over several metabolic events such as glycemia and lacticemia. Amylin has long been shown to induce increases in arterial plasma glucose. We decided to investigate whether amylin plays additional roles in the glucose metabolism. We evaluated glucose homeostasis using whole blood from the tail tip of fasting, conscious, unrestrained normal and streptozotocyn-induced diabetic mice following subcutaneous administration of mouse amylin. Subcutaneous injection of 1 µg mouse amylin caused a transient decrease in whole blood glucose in both normal and diabetic mice in the absence of insulin. The blood glucose levels were lowest approximately 2 hours after amylin administration, after that they gradually recovered to the levels of the control group. The hypoglycemic effect followed a dose-dependent response ranging from 0.1 to 50 µg / mouse. These results reveal the ability for amylin in the direct control of glycemia at low doses in the absence of insulin.


Subject(s)
Hypoglycemia/chemically induced , Islet Amyloid Polypeptide/pharmacology , Animals , Diabetes Mellitus, Experimental , Dose-Response Relationship, Drug , Fasting/metabolism , Homeostasis , Islet Amyloid Polypeptide/administration & dosage , Male , Mice , Pancreas/metabolism
18.
Arterioscler Thromb Vasc Biol ; 32(9): 2185-98, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22796577

ABSTRACT

OBJECTIVE: Blood-sucking arthropods' salivary glands contain a remarkable diversity of antihemostatics. The aim of the present study was to identify the unique salivary anticoagulant of the sand fly Lutzomyia longipalpis, which remained elusive for decades. METHODS AND RESULTS: Several L. longipalpis salivary proteins were expressed in human embryonic kidney 293 cells and screened for inhibition of blood coagulation. A novel 32.4-kDa molecule, named Lufaxin, was identified as a slow, tight, noncompetitive, and reversible inhibitor of factor Xa (FXa). Notably, Lufaxin's primary sequence does not share similarity to any physiological or salivary inhibitors of coagulation reported to date. Lufaxin is specific for FXa and does not interact with FX, Dansyl-Glu-Gly-Arg-FXa, or 15 other enzymes. In addition, Lufaxin blocks prothrombinase and increases both prothrombin time and activated partial thromboplastin time. Surface plasmon resonance experiments revealed that FXa binds Lufaxin with an equilibrium constant ≈3 nM, and isothermal titration calorimetry determined a stoichiometry of 1:1. Lufaxin also prevents protease-activated receptor 2 activation by FXa in the MDA-MB-231 cell line and abrogates edema formation triggered by injection of FXa in the paw of mice. Moreover, Lufaxin prevents FeCl(3)-induced carotid artery thrombus formation and prolongs activated partial thromboplastin time ex vivo, implying that it works as an anticoagulant in vivo. Finally, salivary gland of sand flies was found to inhibit FXa and to interact with the enzyme. CONCLUSIONS: Lufaxin belongs to a novel family of slow-tight FXa inhibitors, which display antithrombotic and anti-inflammatory activities. It is a useful tool to understand FXa structural features and its role in prohemostatic and proinflammatory events.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Factor Xa Inhibitors , Fibrinolytic Agents/pharmacology , Inflammation/prevention & control , Insect Proteins/pharmacology , Psychodidae/chemistry , Receptor, PAR-2/antagonists & inhibitors , Salivary Glands/chemistry , Thrombosis/prevention & control , Amino Acid Sequence , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/isolation & purification , Blood Coagulation/drug effects , Calorimetry , Cell Line, Tumor , Chlorides , Cloning, Molecular , Disease Models, Animal , Dose-Response Relationship, Drug , Factor Xa/metabolism , Female , Ferric Compounds , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/isolation & purification , HEK293 Cells , Humans , Inflammation/blood , Inflammation/metabolism , Insect Proteins/chemistry , Insect Proteins/isolation & purification , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Molecular Weight , Partial Thromboplastin Time , Protein Binding , Prothrombin Time , Rats , Receptor, PAR-2/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Surface Plasmon Resonance , Thromboplastin/antagonists & inhibitors , Thromboplastin/metabolism , Thrombosis/blood , Thrombosis/chemically induced , Thrombosis/metabolism , Time Factors
19.
J Biol Chem ; 286(32): 27998-8010, 2011 Aug 12.
Article in English | MEDLINE | ID: mdl-21673107

ABSTRACT

The molecular mechanism of factor Xa (FXa) inhibition by Alboserpin, the major salivary gland anticoagulant from the mosquito and yellow fever vector Aedes albopictus, has been characterized. cDNA of Alboserpin predicts a 45-kDa protein that belongs to the serpin family of protease inhibitors. Recombinant Alboserpin displays stoichiometric, competitive, reversible and tight binding to FXa (picomolar range). Binding is highly specific and is not detectable for FX, catalytic site-blocked FXa, thrombin, and 12 other enzymes. Alboserpin displays high affinity binding to heparin (K(D) ~ 20 nM), but no change in FXa inhibition was observed in the presence of the cofactor, implying that bridging mechanisms did not take place. Notably, Alboserpin was also found to interact with phosphatidylcholine and phosphatidylethanolamine but not with phosphatidylserine. Further, annexin V (in the absence of Ca(2+)) or heparin outcompetes Alboserpin for binding to phospholipid vesicles, suggesting a common binding site. Consistent with its activity, Alboserpin blocks prothrombinase activity and increases both prothrombin time and activated partial thromboplastin time in vitro or ex vivo. Furthermore, Alboserpin prevents thrombus formation provoked by ferric chloride injury of the carotid artery and increases bleeding in a dose-dependent manner. Alboserpin emerges as an atypical serpin that targets FXa and displays unique phospholipid specificity. It conceivably uses heparin and phosphatidylcholine/phosphatidylethanolamine as anchors to increase protein localization and effective concentration at sites of injury, cell activation, or inflammation.


Subject(s)
Aedes/chemistry , Factor Xa Inhibitors , Fibrinolytic Agents/chemistry , Heparin/chemistry , Insect Proteins/chemistry , Insect Vectors/chemistry , Phosphatidylcholines/chemistry , Phosphatidylethanolamines/chemistry , Serpins/chemistry , Yellow Fever , Aedes/genetics , Aedes/metabolism , Aedes/virology , Animals , Blood Coagulation , Factor Xa/chemistry , Factor Xa/genetics , Factor Xa/metabolism , Fibrinolytic Agents/metabolism , Heparin/genetics , Heparin/metabolism , Humans , Insect Proteins/genetics , Insect Proteins/metabolism , Insect Vectors/metabolism , Insect Vectors/virology , Mice , Phosphatidylcholines/genetics , Phosphatidylcholines/metabolism , Phosphatidylethanolamines/genetics , Phosphatidylethanolamines/metabolism , Phosphatidylserines/chemistry , Phosphatidylserines/genetics , Phosphatidylserines/metabolism , Protein Binding , Serpins/genetics , Serpins/metabolism
20.
Thromb Haemost ; 104(6): 1116-23, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20838739

ABSTRACT

Nitrophorin 2 (NP2) is a 20 kDa lipocalin identified in the salivary gland of the blood sucking insect, Rhodnius prolixus. It functions as a potent inhibitor of the intrinsic pathway of coagulation upon binding to factor IX (FIX) or FIXa. Herein we have investigated the in vivo antithrombotic properties of NP2. Surface plasmon resonance assays demonstrated that NP2 binds to rat FIX and FIXa with high affinities (KD = 43 and 47 nM, respectively), and prolongs the aPTT without affecting the PT. In order to evaluate NP2 antithrombotic effects in vivo two distinct models of thrombosis in rats were carried out. In the rose Bengal/laser induced injury model of arterial thrombosis, NP2 increased the carotid artery occlusion time by ≍35 and ≍155%, at doses of 8 and 80 µg/kg, respectively. NP2 also inhibited thrombus formation in an arterio-venous shunt model, showing ≍60% reduction at 400 µg/kg (i.v. administration). The antithrombotic effect lasted for up to 48 hours after a single i.v. dose. Notably, effective doses of NP2 did not increase the blood loss as evaluated by tail-transection model. In conclusion, NP2 is a potent and long-lasting inhibitor of arterial thrombosis with minor effects on haemostasis. It might be regarded as a potential agent for the treatment of human cardiovascular diseases.


Subject(s)
Anticoagulants/pharmacology , Factor IXa/antagonists & inhibitors , Fibrinolytic Agents/pharmacology , Hemeproteins/pharmacology , Hemostasis/drug effects , Salivary Proteins and Peptides/pharmacology , Thrombosis/prevention & control , Animals , Anticoagulants/administration & dosage , Anticoagulants/metabolism , Disease Models, Animal , Factor IXa/metabolism , Female , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/metabolism , Hemeproteins/administration & dosage , Hemeproteins/metabolism , Injections, Intravenous , Kinetics , Male , Partial Thromboplastin Time , Protein Binding , Prothrombin Time , Rats , Rats, Wistar , Salivary Proteins and Peptides/administration & dosage , Salivary Proteins and Peptides/metabolism , Surface Plasmon Resonance , Thrombin/metabolism , Thrombosis/blood , Thrombosis/etiology
SELECTION OF CITATIONS
SEARCH DETAIL
...