Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 26
1.
J Am Chem Soc ; 143(44): 18481-18489, 2021 11 10.
Article En | MEDLINE | ID: mdl-34723512

Cyclotides are plant-derived peptides with complex structures shaped by their head-to-tail cyclic backbone and cystine knot core. These structural features underpin the native bioactivities of cyclotides, as well as their beneficial properties as pharmaceutical leads, including high proteolytic stability and cell permeability. However, their inherent structural complexity presents a challenge for cyclotide engineering, particularly for accessing libraries of sufficient chemical diversity to design potent and selective cyclotide variants. Here, we report a strategy using mRNA display enabling us to select potent cyclotide-based FXIIa inhibitors from a library comprising more than 1012 members based on the cyclotide scaffold of Momordica cochinchinensis trypsin inhibitor-II (MCoTI-II). The most potent and selective inhibitor, cMCoFx1, has a pM inhibitory constant toward FXIIa with greater than three orders of magnitude selectivity over related serine proteases, realizing specific inhibition of the intrinsic coagulation pathway. The cocrystal structure of cMCoFx1 and FXIIa revealed interactions at several positions across the contact interface that conveyed high affinity binding, highlighting that such cyclotides are attractive cystine knot scaffolds for therapeutic development.


Blood Proteins/pharmacology , Cyclotides/pharmacology , Factor XIIa/metabolism , Blood Proteins/chemistry , Cyclotides/chemistry , Factor XIIa/genetics , Gene Expression Regulation/drug effects , Humans
2.
Molecules ; 24(11)2019 Jun 06.
Article En | MEDLINE | ID: mdl-31174390

Pentamidine is bis-oxybenzamidine-based antiprotozoal drug. The parenteral use of pentamidine appears to affect the processes of blood coagulation and/or fibrinolysis resulting in rare but potentially life-threatening blood clot formation. Pentamidine was also found to cause disseminated intravascular coagulation syndrome. To investigate the potential underlying molecular mechanism(s) of pentamidine's effects on coagulation and fibrinolysis, we studied its effects on clotting times in normal and deficient human plasmas. Using normal plasma, pentamidine isethionate doubled the activated partial thromboplastin time at 27.5 µM, doubled the prothrombin time at 45.7 µM, and weakly doubled the thrombin time at 158.17 µM. Using plasmas deficient of factors VIIa, IXa, XIa, or XIIa, the concentrations to double the activated partial thromboplastin time were similar to that obtained using normal plasma. Pentamidine also inhibited plasmin-mediated clot lysis with half-maximal inhibitory concentration (IC50) value of ~3.6 µM. Chromogenic substrate hydrolysis assays indicated that pentamidine inhibits factor Xa and plasmin with IC50 values of 10.4 µM and 8.4 µM, respectively. Interestingly, it did not significantly inhibit thrombin, factor XIa, factor XIIIa, neutrophil elastase, or chymotrypsin at the highest concentrations tested. Michaelis-Menten kinetics and molecular modeling studies revealed that pentamidine inhibits factor Xa and plasmin in a competitive fashion. Overall, this study provides quantitative mechanistic insights into the in vitro effects of pentamidine isethionate on coagulation and fibrinolysis via the disruption of the proteolytic activity of factor Xa and plasmin.


Blood Coagulation/drug effects , Fibrinolysis/drug effects , Pentamidine/pharmacology , Thrombosis/drug therapy , Blood Coagulation Tests , Factor VIIa/genetics , Factor XIIa/genetics , Factor XIa/genetics , Factor Xa/genetics , Humans , Partial Thromboplastin Time , Prothrombin Time , Thrombin/chemistry , Thrombin/genetics , Thrombin Time , Thrombosis/blood , Thrombosis/pathology
3.
J Thromb Haemost ; 17(1): 183-194, 2019 01.
Article En | MEDLINE | ID: mdl-30394658

Essentials During contact system activation, factor XII is progressively cleaved by plasma kallikrein. We investigated the role of factor XII truncation in biochemical studies. Factor XII contains naturally occurring truncating cleavage sites for a variety of enzymes. Truncation of factor XII primes it for activation in solution through exposure of R353. SUMMARY: Background The contact activation system and innate immune system are interlinked in inflammatory pathology. Plasma kallikrein (PKa) is held responsible for the stepwise processing of factor XII (FXII). A first cleavage activates FXII (into FXIIa); subsequent cleavages truncate it. This truncation eliminates its surface-binding domains, which negatively regulates surface-dependent coagulation. Objectives To investigate the influence of FXII truncation on its activation and downstream kallikrein-kinin system activation. Methods We study activation of recombinant FXII variants by chromogenic assays, by FXIIa ELISA and western blotting. Results We demonstrate that FXII truncation primes it for activation by PKa in solution. We demonstrate this phenomenon in three settings. (i) Truncation at a naturally occurring PKa-sensitive cleavage site, R334, accelerates FXIIa formation in solution. A site-directed mutant FXII-R334A displays ~50% reduced activity when exposed to PKa. (ii) A pathogenic mutation in FXII that causes hereditary angioedema, introduces an additional plasmin-sensitive cleavage site. Truncation at this site synergistically accelerates FXII activation in solution. (iii) We identify new, naturally occurring cleavage sites in FXII that have so far not been functionally linked to contact system activation. As examples, we show that non-activating truncation of FXII by neutrophil elastase and cathepsin K primes it for activation by PKa in solution. Conclusions FXII truncation, mediated by either pathogenic mutations or naturally occurring cleavage sites, primes FXII for activation in solution. We propose that the surface-binding domains of FXII shield its activating cleavage site, R353. This may help to explain how the contact system contributes to inflammatory pathology.


Blood Coagulation , Factor XII/metabolism , Factor XIIa/metabolism , Plasma Kallikrein/metabolism , Cathepsin K/metabolism , Enzyme Activation , Factor XII/genetics , Factor XIIa/genetics , HEK293 Cells , Humans , Leukocyte Elastase/metabolism , Mutation , Proline-Rich Protein Domains , Protein Interaction Domains and Motifs , Substrate Specificity , Time Factors
4.
Arterioscler Thromb Vasc Biol ; 38(8): 1748-1760, 2018 08.
Article En | MEDLINE | ID: mdl-30354195

Objective- Terminal complications of bacterial sepsis include development of disseminated intravascular consumptive coagulopathy. Bacterial constituents, including long-chain polyphosphates (polyP), have been shown to activate the contact pathway of coagulation in plasma. Recent work shows that activation of the contact pathway in flowing whole blood promotes thrombin generation and platelet activation and consumption distal to thrombus formation ex vivo and in vivo. Here, we sought to determine whether presence of long-chain polyP or bacteria in the bloodstream promotes platelet activation and consumption in a coagulation factor (F)XII-dependent manner. Approach and Results- Long-chain polyP promoted platelet P-selectin expression, microaggregate formation, and platelet consumption in flowing whole blood in a contact activation pathway-dependent manner. Moreover, long-chain polyP promoted local fibrin formation on collagen under shear flow in a FXI-dependent manner. Distal to the site of thrombus formation, platelet consumption was dramatically enhanced in the presence of long-chain polyP in the blood flow in a FXI- and FXII-dependent manner. In a murine model, long-chain polyP promoted platelet deposition and fibrin generation in lungs in a FXII-dependent manner. In a nonhuman primate model of bacterial sepsis, pre-treatment of animals with an antibody blocking FXI activation by FXIIa reduced lethal dose100 Staphylococcus aureus-induced platelet and fibrinogen consumption. Conclusions- This study demonstrates that bacterial-type long-chain polyP promotes platelet activation in a FXII-dependent manner in flowing blood, which may contribute to sepsis-associated thrombotic processes, consumptive coagulopathy, and thrombocytopenia.


Blood Coagulation/drug effects , Blood Platelets/drug effects , Factor XII/metabolism , Factor XIIa/metabolism , Platelet Activation/drug effects , Polyphosphates/toxicity , Thrombosis/chemically induced , Animals , Blood Platelets/metabolism , Disease Models, Animal , Factor XII/genetics , Factor XIIa/genetics , Female , Fibrin/metabolism , Humans , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Papio ursinus , Prekallikrein/genetics , Prekallikrein/metabolism , Pulmonary Embolism/blood , Pulmonary Embolism/chemically induced , Pulmonary Embolism/genetics , Sepsis/blood , Sepsis/microbiology , Signal Transduction/drug effects , Staphylococcal Infections/blood , Staphylococcal Infections/microbiology , Thrombosis/blood , Thrombosis/genetics , Tissue Kallikreins/genetics , Tissue Kallikreins/metabolism
5.
Biochim Biophys Acta Mol Cell Res ; 1864(11 Pt B): 2118-2127, 2017 Nov.
Article En | MEDLINE | ID: mdl-28743596

The contact system is a potent procoagulant and proinflammatory plasma protease cascade that is initiated by binding ("contact")-induced, auto-activation of factor XII zymogen. Formed active serine protease FXIIa then cleaves plasma prekallikrein to kallikrein that in turn liberates the mediator bradykinin from its precursor high molecular weight kininogen. Bradykinin induces inflammation with implications for host defense and innate immunity. FXIIa also triggers the intrinsic pathway of coagulation that has been shown to critically contribute to thrombosis. Vice versa, FXII deficiency impairs thrombosis in animal models without inducing abnormal excessive bleeding. Recent work has established the FXIIa-driven contact system as promising target for anticoagulant and anti-inflammatory drugs. This review focuses on the biochemistry of the contact system, its regulation by endogenous and exogenous inhibitors, and roles in disease states. This article is part of a Special Issue entitled: Proteolysis as a Regulatory Event in Pathophysiology edited by Stefan Rose-John.


Blood Coagulation/genetics , Factor XII Deficiency/genetics , Factor XIIa/genetics , Inflammation/genetics , Bradykinin/genetics , Factor XII Deficiency/blood , Factor XII Deficiency/pathology , Humans , Immunity, Innate/genetics , Inflammation/blood , Inflammation/pathology , Kallikreins/genetics , Thrombosis/blood , Thrombosis/genetics , Thrombosis/pathology
6.
Curr Opin Hematol ; 24(5): 411-418, 2017 Sep.
Article En | MEDLINE | ID: mdl-28604413

PURPOSE OF REVIEW: Exposure of blood to foreign surfaces induces reciprocal conversion of the plasma proteins factor XII (fXII) and plasma prekallikrein (PPK) to the proteases α-fXIIa and α-kallikrein. This process, called contact activation, has a range of effects on host defence mechanisms, including promoting coagulation. The nature of the triggering mechanism for contact activation is debated. One hypothesis predicts that fXII has protease activity, either intrinsically or upon surface-binding, that initiates contact activation. We tested this by assessing the proteolytic activity of a recombinant fXII variant that cannot be converted to α-fXIIa. RECENT FINDINGS: The proteolytic activity of fXII-T (for 'triple' mutant), a variant with alanine substitutions for arginine at activation cleavage sites (Arg334, Arg344, and Arg353) was tested with known α-fXIIa substrates. FXII-T activates PPK in solution, and the reaction is enhanced by polyphosphate, an inducer of contact activation released from platelets. In the presence of polyphosphate, fXII-T converts fXII to α-fXIIa, and also converts the coagulation protein factor XI to its active form. SUMMARY: The findings support the hypothesis that contact activation is initiated through activity intrinsic to single-chain fXII, and indicate that preexisting α-fXIIa is not required for induction of contact activation.


Factor XIIa/metabolism , Plasma Kallikrein/metabolism , Prekallikrein/metabolism , Factor XIIa/genetics , Humans , Plasma Kallikrein/genetics , Prekallikrein/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
7.
Semin Thromb Hemost ; 43(8): 814-826, 2017 Nov.
Article En | MEDLINE | ID: mdl-28346966

Contact activation is the surface-induced conversion of factor XII (FXII) zymogen to the serine protease FXIIa. Blood-circulating FXII binds to negatively charged surfaces and this contact to surfaces triggers a conformational change in the zymogen inducing autoactivation. Several surfaces that have the capacity for initiating FXII contact activation have been identified, including misfolded protein aggregates, collagen, nucleic acids, and platelet and microbial polyphosphate. Activated FXII initiates the proinflammatory kallikrein-kinin system and the intrinsic coagulation pathway, leading to formation of bradykinin and thrombin, respectively. FXII contact activation is well characterized in vitro and provides the mechanistic basis for the diagnostic clotting assay, activated partial thromboplastin time. However, only in the past decade has the critical role of FXII contact activation in pathological thrombosis been appreciated. While defective FXII contact activation provides thromboprotection, excess activation underlies the swelling disorder hereditary angioedema type III. This review provides an overview of the molecular basis of FXII contact activation and FXII contact activation-associated disease states.


Blood Coagulation , Bradykinin/metabolism , Factor XIIa/metabolism , Hereditary Angioedema Type III/metabolism , Thrombin/metabolism , Animals , Bradykinin/genetics , Enzyme Activation , Factor XIIa/genetics , Hereditary Angioedema Type III/genetics , Humans , Thrombin/genetics
8.
J Neuroinflammation ; 14(1): 39, 2017 02 20.
Article En | MEDLINE | ID: mdl-28219400

BACKGROUND: Traumatic brain injury (TBI) is a devastating neurological condition and a frequent cause of permanent disability. Posttraumatic inflammation and brain edema formation, two pathological key events contributing to secondary brain injury, are mediated by the contact-kinin system. Activation of this pathway in the plasma is triggered by activated factor XII. Hence, we set out to study in detail the influence of activated factor XII on the abovementioned pathophysiological features of TBI. METHODS: Using a cortical cryogenic lesion model in mice, we investigated the impact of genetic deficiency of factor XII and inhibition of activated factor XII with a single bolus injection of recombinant human albumin-fused Infestin-4 on the release of bradykinin, the brain lesion size, and contact-kinin system-dependent pathological events. We determined protein levels of bradykinin, intracellular adhesion molecule-1, CC-chemokine ligand 2, and interleukin-1ß by enzyme-linked immunosorbent assays and mRNA levels of genes related to inflammation by quantitative real-time PCR. Brain lesion size was determined by tetrazolium chloride staining. Furthermore, protein levels of the tight junction protein occludin, integrity of the blood-brain barrier, and brain water content were assessed by Western blot analysis, extravasated Evans Blue dye, and the wet weight-dry weight method, respectively. Infiltration of neutrophils and microglia/activated macrophages into the injured brain lesions was quantified by immunohistological stainings. RESULTS: We show that both genetic deficiency of factor XII and inhibition of activated factor XII in mice diminish brain injury-induced bradykinin release by the contact-kinin system and minimize brain lesion size, blood-brain barrier leakage, brain edema formation, and inflammation in our brain injury model. CONCLUSIONS: Stimulation of bradykinin release by activated factor XII probably plays a prominent role in expanding secondary brain damage by promoting brain edema formation and inflammation. Pharmacological blocking of activated factor XII could be a useful therapeutic principle in the treatment of TBI-associated pathologic processes by alleviating posttraumatic inflammation and brain edema formation.


Brain Edema/metabolism , Brain Edema/prevention & control , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/prevention & control , Factor XIIa/antagonists & inhibitors , Factor XIIa/metabolism , Animals , Bradykinin/metabolism , Brain Injuries/metabolism , Brain Injuries/prevention & control , Factor XIIa/genetics , Inflammation/metabolism , Inflammation/prevention & control , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
9.
Thromb Haemost ; 117(1): 176-187, 2017 01 05.
Article En | MEDLINE | ID: mdl-27786338

Atherosclerosis is considered a chronic inflammatory disease of the vessel wall. Coagulation pathways and immune responses contribute to disease development. The role of coagulation factor XII (FXII) in vascular inflammation, however, remains controversial. We here investigated the function of FXII in atherosclerosis using apolipoprotein E and FXII-deficient (F12-/-Apoe-/-) mice. Compared to F12+/+Apoe-/- controls, atherosclerotic lesion formation was reduced in F12-/-Apoe-/- mice. This was associated with a decrease in serum interleukin (IL)-1ß and IL-12 levels and reduced expression of pro-inflammatory cytokines in the aorta in atherosclerotic F12-/-Apoe-/- mice, as well as diminished Th1-cell differentiation in the aorta, blood, and lymphoid organs. No changes in circulating bradykinin, thrombin-antithrombin-complexes or plasminogen were observed. Mechanistically, activated FXII (FXIIa) was revealed to directly induce bone marrow-derived macrophages to secrete pro-inflammatory cytokines, including tumour necrosis factor-α, IL-1ß, IL-12, and IL-6. Exposure of bone marrow-derived antigen presenting cells to FXIIa similarly induced pro-inflammatory cytokines, and an enhanced capacity to trigger antigen-specific interferon γ-production in CD4+ T cells. Notably, bone-marrow derived macrophages were capable of directly activating FXII. Moreover, the induction of cytokine expression by FXIIa in macrophages occurred independently of FXII protease enzymatic activity and was decreased upon phospholipase C treatment, suggesting urokinase-type plasminogen activator receptor (uPAR) to confer FXIIa-induced cell signalling. These data reveal FXII to play an important role in atherosclerotic lesion formation by functioning as a strong inducer of pro-inflammatory cytokines in antigen-presenting cells. Targeting of FXII may thus be a promising approach for treating cardiovascular disease.


Antigen-Presenting Cells/metabolism , Aortic Diseases/metabolism , Atherosclerosis/metabolism , Cytokines/metabolism , Factor XII Deficiency/metabolism , Factor XII/metabolism , Inflammation Mediators/metabolism , Macrophages/metabolism , Animals , Antigen-Presenting Cells/immunology , Aortic Diseases/blood , Aortic Diseases/genetics , Aortic Diseases/immunology , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/immunology , Cell Proliferation , Cytokines/immunology , Disease Models, Animal , Factor XII/genetics , Factor XII Deficiency/blood , Factor XII Deficiency/genetics , Factor XII Deficiency/immunology , Factor XIIa/genetics , Factor XIIa/metabolism , Genetic Predisposition to Disease , Inflammation Mediators/immunology , Lymphocyte Activation , Macrophages/immunology , Mice, Inbred C57BL , Mice, Knockout, ApoE , Phenotype , Plaque, Atherosclerotic , Receptors, Urokinase Plasminogen Activator/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Time Factors
10.
BMC Med Genet ; 17: 21, 2016 Mar 11.
Article En | MEDLINE | ID: mdl-26969407

BACKGROUND: Plasma coagulation Factor XIIa (Hageman factor; encoded by F12) and kallikrein (KAL or Fletcher factor; encoded by KLKB1) are proteases of the kallikerin-kinin system involved in converting the inactive circulating prorenin to renin. Renin is a key enzyme in the formation of angiotensin II, which regulates blood pressure, fluid and electrolyte balance and is a biomarker for cardiovascular, metabolic and renal function. The renin-angiotensin system is implicated in extinction learning in posttraumatic stress disorder. METHODS & RESULTS: Active plasma renin was measured from two independent cohorts- civilian twins and siblings, as well as U.S. Marines, for a total of 1,180 subjects. Genotyping these subjects revealed that the carriers of the minor alleles at the two loci- F12 and KLKB1 had a significant association with reduced levels of active plasma renin. Meta-analyses confirmed the association across cohorts. In vitro studies verified digestion of human recombinant pro-renin by kallikrein (KAL) to generate active renin. Subsequently, the active renin was able to digest the synthetic substrate angiotensinogen to angiotensin-I. Examination of mouse juxtaglomerular cell line and mouse kidney sections showed co-localization of KAL with renin. Expression of either REN or KLKB1 was regulated in cell line and rodent models of hypertension in response to oxidative stress, interleukin or arterial blood pressure changes. CONCLUSIONS: The functional variants of KLKB1 (rs3733402) and F12 (rs1801020) disrupted the cascade of enzymatic events, resulting in diminished formation of active renin. Using genetic, cellular and molecular approaches we found that conversion of zymogen prorenin to renin was influenced by these polymorphisms. The study suggests that the variant version of protease factor XIIa due to the amino acid substitution had reduced ability to activate prekallikrein to KAL. As a result KAL has reduced efficacy in converting prorenin to renin and this step of the pathway leading to activation of renin affords a potential therapeutic target.


Factor XIIa/genetics , Kallikreins/genetics , Polymorphism, Single Nucleotide , Renin-Angiotensin System/genetics , Renin/blood , Adolescent , Adult , Aged , Alleles , Angiotensin I/blood , Angiotensinogen/blood , Animals , Blood Pressure , Cell Cycle Proteins , Cell Line , Gene Expression Regulation , Genetic Loci , Genome-Wide Association Study , Genotyping Techniques , Humans , Hypertension/genetics , Juxtaglomerular Apparatus/cytology , Kallikreins/blood , Male , Mice , Middle Aged , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Prekallikrein/metabolism , Renin/genetics , Serine Endopeptidases/metabolism , Transferases , Young Adult
11.
Proc Natl Acad Sci U S A ; 112(13): 4068-73, 2015 Mar 31.
Article En | MEDLINE | ID: mdl-25775543

Alzheimer's disease (AD) is characterized by accumulation of the ß-amyloid peptide (Aß), which likely contributes to disease via multiple mechanisms. Increasing evidence implicates inflammation in AD, the origins of which are not completely understood. We investigated whether circulating Aß could initiate inflammation in AD via the plasma contact activation system. This proteolytic cascade is triggered by the activation of the plasma protein factor XII (FXII) and leads to kallikrein-mediated cleavage of high molecular-weight kininogen (HK) and release of proinflammatory bradykinin. Aß has been shown to promote FXII-dependent cleavage of HK in vitro. In addition, increased cleavage of HK has been found in the cerebrospinal fluid of patients with AD. Here, we show increased activation of FXII, kallikrein activity, and HK cleavage in AD patient plasma. Increased contact system activation is also observed in AD mouse model plasma and in plasma from wild-type mice i.v. injected with Aß42. Our results demonstrate that Aß42-mediated contact system activation can occur in the AD circulation and suggest new pathogenic mechanisms, diagnostic tests, and therapies for AD.


Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Factor XII/metabolism , Factor XIIa/metabolism , Aged , Aged, 80 and over , Amyloid beta-Peptides/metabolism , Animals , Biomarkers/metabolism , Case-Control Studies , Dementia/genetics , Dementia/metabolism , Disease Models, Animal , Disease Progression , Factor XII/genetics , Factor XIIa/genetics , Female , Humans , Inflammation , Kallikreins/blood , Kininogens/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission , Peptide Fragments/metabolism , Transferrin/metabolism
12.
Haematologica ; 100(3): 308-14, 2015 Mar.
Article En | MEDLINE | ID: mdl-25596265

An excess of free heme is present in the blood during many types of hemolytic anemia. This has been linked to organ damage caused by heme-mediated oxidative stress and vascular inflammation. We investigated the mechanism of heme-induced coagulation activation in vivo. Heme caused coagulation activation in wild-type mice that was attenuated by an anti-tissue factor antibody and in mice expressing low levels of tissue factor. In contrast, neither factor XI deletion nor inhibition of factor XIIa-mediated factor XI activation reduced heme-induced coagulation activation, suggesting that the intrinsic coagulation pathway is not involved. We investigated the source of tissue factor in heme-induced coagulation activation. Heme increased the procoagulant activity of mouse macrophages and human PBMCs. Tissue factor-positive staining was observed on leukocytes isolated from the blood of heme-treated mice but not on endothelial cells in the lungs. Furthermore, heme increased vascular permeability in the mouse lungs, kidney and heart. Deletion of tissue factor from either myeloid cells, hematopoietic or endothelial cells, or inhibition of tissue factor expressed by non-hematopoietic cells did not reduce heme-induced coagulation activation. However, heme-induced activation of coagulation was abolished when both non-hematopoietic and hematopoietic cell tissue factor was inhibited. Finally, we demonstrated that coagulation activation was partially attenuated in sickle cell mice treated with recombinant hemopexin to neutralize free heme. Our results indicate that heme promotes tissue factor-dependent coagulation activation and induces tissue factor expression on leukocytes in vivo. We also demonstrated that free heme may contribute to thrombin generation in a mouse model of sickle cell disease.


Anemia, Hemolytic/genetics , Anemia, Sickle Cell/genetics , Blood Coagulation/drug effects , Heme/administration & dosage , Thromboplastin/genetics , Anemia, Hemolytic/blood , Anemia, Hemolytic/chemically induced , Anemia, Hemolytic/pathology , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/pathology , Animals , Antibodies/pharmacology , Blood Coagulation/genetics , Capillary Permeability/drug effects , Cells, Cultured , Factor XI/genetics , Factor XI/metabolism , Factor XIIa/antagonists & inhibitors , Factor XIIa/genetics , Factor XIIa/metabolism , Female , Gene Deletion , Gene Expression , Hemopexin/pharmacology , Humans , Injections, Intravenous , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Swine , Thromboplastin/antagonists & inhibitors , Thromboplastin/metabolism
13.
Oral Dis ; 20(6): 616-23, 2014 Sep.
Article En | MEDLINE | ID: mdl-24112124

OBJECTIVE: Gingival epithelium plays a key role in the protection of oral tissues from microbial challenge, especially during the periodontal disease. This study was aimed to evaluate levels of mRNA transcripts of different forms of transglutaminase in the human gingival tissues from patients with chronic periodontitis and relative controls. SUBJECTS AND METHODS: This study included 22 patients with chronic periodontitis (CP) and 22 healthy controls. For each patient, the values of probing depth (PD), clinical attachment level (CAL), and bleeding on probing (BOP) were recorded. Gene expression of transglutaminase 1, transglutaminase 2, transglutaminase 3, and metalloprotease 2 was evaluated by real-time PCR, while that of Factor XIIIA and metalloprotease 9 by RT-PCR. RESULTS: The values of all the clinical parameters were significantly higher in the CP group than in the healthy control group (P < 0.05). In the CP group, the mRNA expression of transglutaminase 1 and transglutaminase 3 was significantly decreased in comparison with healthy control group. A slight nonsignificant changes of transglutaminase 2 gene expression were observed in samples from CP patients in comparison with controls. CONCLUSIONS: These observations suggest that transglutaminase gene expression may be modified in response to chronic injury in the damaged gingival and emphasizes the key role of these enzymes in gingival remodelling/healing and adaptive processes.


GTP-Binding Proteins/genetics , Gene Expression , Periodontitis/genetics , Transglutaminases/genetics , Case-Control Studies , Chronic Disease , Factor XIIa/genetics , Female , GTP-Binding Proteins/metabolism , Humans , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Periodontitis/metabolism , Protein Glutamine gamma Glutamyltransferase 2 , Transglutaminases/metabolism
14.
PLoS One ; 8(8): e74043, 2013.
Article En | MEDLINE | ID: mdl-24013493

Hereditary angioedema (HAE) is predominantly caused by a deficiency in C1 esterase inhibitor (C1INH) (HAE-C1INH). C1INH inhibits activated factor XII (FXIIa), activated factor XI (FXIa), and kallikrein. In HAE-C1INH patients the thrombotic risk is not increased even though activation of the contact system is poorly regulated. Therefore, we hypothesized that contact activation preferentially leads to kallikrein formation and less to activation of the coagulation cascade in HAE-C1INH patients. We measured the levels of C1INH in complex with activated contact factors in plasma samples of HAE-C1INH patients (N=30, 17 during remission and 13 during acute attack) and healthy controls (N=10). We did not detect differences in enzyme-inhibitor complexes between samples of controls, patients during remission and patients during an acute attack. Reconstitution with C1INH did not change this result. Next, we determined the potential to form enzyme-inhibitory complexes after complete in vitro activation of the plasma samples with a FXII trigger. In all samples, enzyme-C1INH levels increased after activation even in patients during an acute attack. However, the levels of FXIIa-C1INH, FXIa-C1INH and kallikrein-C1INH were at least 52% lower in samples taken during remission and 70% lower in samples taken during attack compared to samples from controls (p<0.05). Addition of C1INH after activation led to an increase in levels of FXIIa-C1INH and FXIa-C1INH (p<0.05), which were still lower than in controls (p<0.05), while the levels of kallikrein-C1INH did not change. These results are consistent with constitutive activation and attenuated depletion of the contact system and show that the ongoing activation of the contact system, which is present in HAE-C1INH patients both during remission and during acute attacks, is not associated with preferential generation of kallikrein over FXIa.


Angioedemas, Hereditary/blood , Complement C1 Inhibitor Protein/metabolism , Factor XIIa/metabolism , Factor XIa/metabolism , Kallikreins/blood , Multiprotein Complexes/blood , Adult , Aged , Angioedemas, Hereditary/genetics , Complement C1 Inhibitor Protein/genetics , Factor XIIa/genetics , Factor XIa/genetics , Female , Humans , Kallikreins/genetics , Male , Middle Aged , Multiprotein Complexes/genetics
15.
Circulation ; 121(13): 1510-7, 2010 Apr 06.
Article En | MEDLINE | ID: mdl-20308613

BACKGROUND: Blood coagulation is a tightly regulated process of sequentially activated serine proteases culminating in fibrin formation, which is critical for limiting posttraumatic blood loss but also may contribute to acute thrombotic diseases, most notably myocardial infarction and stroke. Recent studies with factor XII-deficient mice revealed that the factor XII-induced intrinsic coagulation pathway is essential for pathological thrombus formation but dispensable for hemostasis. Consequently, these findings led to the hypothesis that factor XII could be a promising pharmacological target for safe antithrombotic therapy. METHODS AND RESULTS: The complementary DNA of the previously described factor XIIa inhibitor Infestin-4, cloned from the midgut of Triatoma infestans, was fused to recombinant human albumin (rHA) and analyzed in vitro. The resulting protein rHA-Infestin-4 specifically inhibits factor XIIa and causes prolonged activated partial thromboplastin time in human, mouse, and rat plasma. To assess its inhibitory potency in vivo, mice and rats were injected with rHA-Infestin-4 and challenged in pathological thrombus formation models. In addition, bleeding assays were performed. rHA-Infestin-4 completely abolished occlusive arterial thrombus formation in mice and rats while leaving hemostasis fully intact. Furthermore, rHA-Infestin-4 was highly protective in a murine model of ischemic stroke. CONCLUSIONS: These results identify rHA-Infestin-4 as a promising agent to achieve powerful protection from ischemic cardiovascular and cerebrovascular events without affecting hemostasis.


Albumins/pharmacology , Arterial Occlusive Diseases/drug therapy , Factor XIIa/antagonists & inhibitors , Infarction, Middle Cerebral Artery/drug therapy , Insect Proteins/pharmacology , Thrombosis/drug therapy , Albumins/genetics , Animals , Arterial Occlusive Diseases/blood , Brain Ischemia/blood , Brain Ischemia/drug therapy , DNA, Complementary , Factor XIIa/genetics , Factor XIIa/metabolism , Hemostasis/drug effects , Humans , Infarction, Middle Cerebral Artery/blood , Insect Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Partial Thromboplastin Time , Rats , Rats, Inbred Strains , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Thrombosis/blood
16.
Blood Coagul Fibrinolysis ; 20(5): 325-32, 2009 Jul.
Article En | MEDLINE | ID: mdl-19474702

In a subgroup of hereditary angioedema (HAE) patients with normal C1-esterase inhibitor levels, HAE is caused by a Thr309Lys mutation in the coagulation factor XII (F12) gene. The aim of this study was to examine elements of the kallikrein-kinin system ('contact system') and the downstream-linked coagulation, complement and fibrinolytic systems in the plasma of six patients with HAE caused by the Thr309Lys mutation and healthy probands. Blood samples were taken from participants during the symptom-free interval between attacks. Samples were analyzed for activity and concentrations of components of the kallikrein-kinin system and linked enzyme systems. The mean FXII clotting activity was 90% in patients with the FXII mutation, and the concentration of FXIIa was 4.1 ng/ml; this did not differ from healthy probands. Mean prekallikrein amidolytic activity and high-molecular-weight kininogen clotting activity were 130 and 144%, respectively, both higher than in healthy probands. The mean kallikrein-like activity of the HAE patients was 11.4 U/l and did not differ from healthy probands. There was no difference in FXII surface activation by silicon dioxide or in kallikrein-like activity with and without activation by dextran sulfate. Contrary to the results of a recently published study, no indication that the Thr309Lys mutation causes a 'gain-of-function' of FXIIa was observed in this investigation.


Amino Acid Substitution , Angioedemas, Hereditary/genetics , Factor XIIa/genetics , Fibrinolysis/genetics , Kallikreins/blood , Kinins/blood , Mutation, Missense , Point Mutation , Adolescent , Adult , Aged , Aged, 80 and over , Angioedemas, Hereditary/blood , Blood Proteins/analysis , Chromogenic Compounds/metabolism , Complement Pathway, Classical/genetics , Dextran Sulfate/pharmacology , Enzyme Activation , Factor XIIa/physiology , Female , Humans , Kallikreins/antagonists & inhibitors , Male , Middle Aged , Prekallikrein/analysis , Silicon Dioxide/pharmacology , Tissue Plasminogen Activator/blood , Young Adult
18.
J Biol Chem ; 283(27): 18655-64, 2008 Jul 04.
Article En | MEDLINE | ID: mdl-18441012

Coagulation factor XI (FXI) is a covalent homodimer consisting of two identical subunits of 80 kDa linked by a disulfide bond formed by Cys-321 within the Apple 4 domain of each subunit. Because FXI(C321S) is a noncovalent dimer, residues within the interface between the two subunits must mediate its homodimeric structure. The crystal structure of FXI demonstrates formation of salt bridges between Lys-331 of one subunit and Glu-287 of the other subunit and hydrophobic interactions at the interface of the Apple 4 domains involving Ile-290, Leu-284, and Tyr-329. FXI(C321S), FXI(C321S,K331A), FXI(C321S,E287A), FXI(C321S,I290A), FXI(C321S,Y329A), FXI(C321S,L284A), FXI(C321S,K331R), and FXI(C321S,H343A) were expressed in HEK293 cells and characterized using size exclusion chromatography, analytical ultracentrifugation, electron microscopy, and functional assays. Whereas FXI(C321S) and FXI(C321S,H343A) existed in monomer/dimer equilibrium (K(d) approximately 40 nm), all other mutants were predominantly monomers with impaired dimer formation by analytical ultracentrifugation (K(d)=3-38 microm). When converted to the active enzyme, FXIa, all the monomeric mutants activated FIX similarly to wild-type dimeric FXIa. In contrast, these monomeric mutants could not be activated efficiently by FXIIa, thrombin, or autoactivation in the presence of dextran sulfate. We conclude that salt bridges formed between Lys-331 of one subunit and Glu-287 of the other together with hydrophobic interactions at the interface, involving residues Ile-290, Leu-284, and Tyr-329, are essential for homodimer formation. The dimeric structure of FXI is essential for normal proteolytic activation of FXI by FXIIa, thrombin, or FXIa either in solution or on an anionic surface but not for FIX activation by FXIa in solution.


Factor XIIa/chemistry , Factor XI/chemistry , Factor XIa/chemistry , Thrombin/chemistry , Amino Acid Substitution , Crystallography, X-Ray , Dimerization , Disulfides/chemistry , Disulfides/metabolism , Factor XI/genetics , Factor XI/metabolism , Factor XIIa/genetics , Factor XIIa/metabolism , Factor XIa/genetics , Factor XIa/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Mutation, Missense , Protein Structure, Quaternary/genetics , Protein Structure, Tertiary/genetics , Thrombin/genetics , Thrombin/metabolism
19.
Biochemistry ; 46(50): 14450-60, 2007 Dec 18.
Article En | MEDLINE | ID: mdl-18020374

The zymogen, factor XI, and the enzyme, factor XIa, interact specifically with functional receptors on the surface of activated platelets. These studies were initiated to identify the molecular subdomain within factor XIa that binds to activated platelets. Both factor XIa (Ki approximately 1.4 nM) and a chimeric factor XIa containing the Apple 3 domain of prekallikrein (Ki approximately 2.7 nM) competed with [125I]factor XIa for binding sites on activated platelets, suggesting that the factor XIa binding site for platelets is not located in the Apple 3 domain which mediates factor XI binding to platelets. The recombinant catalytic domain (Ile370-Val607) inhibited the binding of [125I]factor XIa to the platelets (Ki approximately 3.5 nM), whereas the recombinant factor XI heavy chain did not, demonstrating that the platelet binding site is located in the light chain of factor XIa. A conformationally constrained cyclic peptide (Cys527-Cys542) containing a high-affinity (KD approximately 86 nM) heparin-binding site within the catalytic domain of factor XIa also displaced [125I]factor XIa from the surface of activated platelets (Ki approximately 5.8 nM), whereas a scrambled peptide of identical composition was without effect, suggesting that the binding site in factor XIa that interacts with the platelet surface resides in the catalytic domain near the heparin binding site of factor XIa. These data support the conclusion that a conformational transition accompanies conversion of factor XI to factor XIa that conceals the Apple 3 domain factor XI (zymogen) platelet binding site and exposes the factor XIa (enzyme) platelet binding site within the catalytic domain possibly comprising residues Cys527-Cys542.


Blood Platelets/metabolism , Factor XIa/chemistry , Factor XIa/metabolism , Amino Acid Sequence , Binding Sites , Catalysis , Cell Line , Cysteine/chemistry , Cysteine/genetics , Cysteine/metabolism , Enzyme-Linked Immunosorbent Assay , Factor XI/chemistry , Factor XI/genetics , Factor XI/metabolism , Factor XIIa/chemistry , Factor XIIa/genetics , Factor XIIa/metabolism , Factor XIa/genetics , Humans , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Prekallikrein/chemistry , Prekallikrein/genetics , Prekallikrein/metabolism , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology, Amino Acid
20.
J Exp Med ; 202(2): 271-81, 2005 Jul 18.
Article En | MEDLINE | ID: mdl-16009717

Blood coagulation is thought to be initiated by plasma protease factor VIIa in complex with the membrane protein tissue factor. In contrast, coagulation factor XII (FXII)-mediated fibrin formation is not believed to play an important role for coagulation in vivo. We used FXII-deficient mice to study the contributions of FXII to thrombus formation in vivo. Intravital fluorescence microscopy and blood flow measurements in three distinct arterial beds revealed a severe defect in the formation and stabilization of platelet-rich occlusive thrombi. Although FXII-deficient mice do not experience spontaneous or excessive injury-related bleeding, they are protected against collagen- and epinephrine-induced thromboembolism. Infusion of human FXII into FXII-null mice restored injury-induced thrombus formation. These unexpected findings change the long-standing concept that the FXII-induced intrinsic coagulation pathway is not important for clotting in vivo. The results establish FXII as essential for thrombus formation, and identify FXII as a novel target for antithrombotic therapy.


Blood Coagulation , Factor XII Deficiency/metabolism , Factor XIIa/metabolism , Thromboembolism/metabolism , Animals , Blood Coagulation/drug effects , Blood Coagulation/genetics , Collagen/administration & dosage , Epinephrine/administration & dosage , Factor VIIa/metabolism , Factor XII Deficiency/genetics , Factor XIIa/genetics , Hemorrhage/genetics , Hemorrhage/metabolism , Lung/metabolism , Lung/pathology , Mice , Mice, Mutant Strains , Platelet Aggregation/drug effects , Platelet Aggregation/genetics , Thromboembolism/chemically induced , Thromboembolism/drug therapy , Thromboembolism/pathology , Vasoconstrictor Agents/administration & dosage
...