Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Alzheimers Dement ; 18(10): 1919-1929, 2022 10.
Article in English | MEDLINE | ID: mdl-34978145

ABSTRACT

Increased activation of the contact system protein high molecular weight kininogen (HK) has been shown in plasma and cerebrospinal fluid of Alzheimer's disease (AD) patients, but its potential role in the brain has not been explored. We assessed HK levels in brain tissue from 20 AD patients and controls and modeled the effects of HK on microglia-like cells in culture. We show increased levels of HK in the hippocampus of AD patients, which colocalized with amyloid beta (Aß) deposits and activated microglia. Treatment of microglia with HK led to cell clustering and elevated levels of phagocytosed Aß. We demonstrate that microglia internalize HK and traffic it to lysosomes, which is accompanied by reduced activity of lysosomal cathepsins L and S. Our results suggest that HK accumulation in the AD hippocampus may alter microglial uptake and degradation of Aß fibrils, possibly contributing to microglial dysfunction in AD.


Subject(s)
Alzheimer Disease , Microglia , Humans , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Cathepsins/metabolism , Cathepsins/pharmacology , Kininogen, High-Molecular-Weight/metabolism , Kininogen, High-Molecular-Weight/pharmacology , Lysosomes/metabolism , Microglia/metabolism , Phagocytosis
2.
Thromb Haemost ; 114(3): 603-13, 2015 Aug 31.
Article in English | MEDLINE | ID: mdl-26063414

ABSTRACT

Cleaved high-molecular-weight kininogen (HKa) or its peptide domain 5 (D5) alone exert anti-adhesive properties in vitro related to impeding integrin-mediated cellular interactions. However, the anti-adhesive effects of HKa in vivo remain elusive. In this study, we investigated the effects of HKa on leukocyte recruitment and neointima formation following wire-induced injury of the femoral artery in C57BL/6 mice. Local application of HKa significantly reduced the accumulation of monocytes and also reduced neointimal lesion size 14 days after injury. Moreover, C57BL/6 mice transplanted with bone marrow from transgenic mice expressing enhanced green fluorescence protein (eGFP) showed a significantly reduced accumulation of eGFP+-cells at the arterial injury site and decreased neointimal lesion size after local application of HKa or the polypeptide D5 alone. A differentiation of accumulating eGFP+-cells into highly specific smooth muscle cells (SMC) was not detected in any group. In contrast, application of HKa significantly reduced the proliferation of locally derived neointimal cells. In vitro, HKa and D5 potently inhibited the adhesion of SMC to vitronectin, thus impairing their proliferation, migration, and survival rates. In conclusion, application of HKa or D5 decreases the inflammatory response to vascular injury and exerts direct effects on SMC by impeding the binding of integrins to extracellular matrix components. Therefore, HKa and D5 may hold promise as novel therapeutic substances to prevent neointima formation.


Subject(s)
Kininogen, High-Molecular-Weight/pharmacology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Neointima , Peptide Fragments/pharmacology , Vascular System Injuries/prevention & control , Animals , Bone Marrow Transplantation , Cell Proliferation/drug effects , Chemotaxis, Leukocyte/drug effects , Disease Models, Animal , Femoral Artery/drug effects , Femoral Artery/injuries , Femoral Artery/metabolism , Femoral Artery/pathology , Genes, Reporter , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Humans , Integrins/metabolism , Kininogen, High-Molecular-Weight/genetics , Kininogen, High-Molecular-Weight/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Structure, Tertiary , Time Factors , U937 Cells , Vascular System Injuries/metabolism , Vascular System Injuries/pathology , Vitronectin/metabolism
3.
Biomaterials ; 37: 1-12, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25443791

ABSTRACT

High-resolution electrophoresis of FXII-derived proteins produced by contact activation of FXII in buffer solutions (i.e. in absence of plasma proteins) with hydrophilic and silanized-glass activators spanning the observable range of water wettability (hydrophilic to hydrophobic), shows no evidence of proteolytic cleavage of FXII into αFXIIa or ßFXIIa. The autoactivation mixture contains only a single-chain protein with a molecular weight of ∼80 kDa, confirming Oscar Ratnoff's previous finding of a single-chain activated form of FXII that he called 'HFea'. Functional assays have shown that these autoactivation products exhibit procoagulant potential (protease activity inducing clotting of blood) or amidolytic potential (cleaves amino bonds in s-2302 chromogen but do not cause coagulation of plasma) or both amidolytic potential and procoagulant potential. Some of these proteins also have the remarkable potential to 'suppress autoactivation' (i.e. suppress creation of enzymes with procoagulant potential). It is thus hypothesized that autoactivation of FXII in the absence of plasma proteins generates not just a single type of activated conformer, as suggested by previous researchers, but rather an ensemble of conformer products with collective activity that varies with activator surface energy used in contact activation of FXII. Furthermore, reaction of αFXIIa with FXII in buffer solution does not produce additional αFXIIa by the putative autoamplification reaction FXIIa + FXII â†’ 2FXIIa as has been proposed in past literature to account for the discrepancy between chromogenic and plasma-coagulation assays for αFXIIa in buffer solution. Instead, net procoagulant activity measured directly by plasma-coagulation assays, decreases systematically with increasing FXII solution concentration. Under the same reaction conditions, chromogenic assay reveals that net amidolytic activity increases with increasing FXII solution concentration. Thus, although autoamplification does not occur it appears that there is some form of "FXII self reaction" that influences products of αFXIIa reaction with FXII. Electrophoretic measurements indicate that no proteolytic cleavage takes in this reaction leading us to conclude that change in activity is most likely due to change(s) in FXII conformation (with related change in enzyme activity).


Subject(s)
Biocompatible Materials/pharmacology , Factor XII/chemistry , Factor XII/metabolism , Hematology , Blood Coagulation/drug effects , Buffers , Electrophoresis , Enzyme Activation/drug effects , Factor XIIa/metabolism , Humans , Hydrolysis/drug effects , Kininogen, High-Molecular-Weight/pharmacology , Prekallikrein/pharmacology , Research , Solutions , Time Factors
4.
Biochem Biophys Res Commun ; 427(3): 497-502, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-23000411

ABSTRACT

High molecular weight kininogen (HK) is a plasma glycoprotein with multiple functions, including the regulation of coagulation. We previously demonstrated that domain 5 (D5(H)), a functional domain of HK, and its derived peptides played an important role in the vitronectin-mediated suppression of cancer cell adhesion and invasion. However, the underlying mechanisms of the D5(H)-mediated suppressive effects remain to be elucidated. Here, we showed that D5(H) and its derivatives inhibited the collagen-mediated cell adhesion and invasion of human osteosarcoma MG63 cells. Using purified D5(H) fused to glutathione-S-transferase (GST) and D5(H)-derived peptides for column chromatography, an actin-binding protein, α-actinin-4, was identified as a binding protein of D5(H) with high-affinity for P-5m, a core octapeptide of D5(H). Immunofluorescence microscopy demonstrated that D5(H) co-localized with α-actinin-4 inside MG63 cells. In addition, exogenous GST-D5(H) added to the culture media was transported into MG63 cells, although GST alone as a control was not. As α-actinin-4 regulates actin polymerization necessary for cell adhesion and is related to the integrin-dependent attachment of cells to the extracellular matrix, our results suggest that D5(H) may modulate cell adhesion and invasion together with actinin-4.


Subject(s)
Actinin/metabolism , Kininogen, High-Molecular-Weight/metabolism , Neoplasms/pathology , Amino Acid Sequence , Cell Adhesion , Cell Line, Tumor , Collagen/metabolism , Humans , Kininogen, High-Molecular-Weight/genetics , Kininogen, High-Molecular-Weight/pharmacology , Molecular Sequence Data , Neoplasm Invasiveness , Neoplasms/metabolism , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Fragments/pharmacology
5.
J Biol Chem ; 286(28): 24561-71, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21586566

ABSTRACT

The nonenzymatic cofactor high molecular weight kininogen (HK) is a precursor of bradykinin (BK). The production of BK from HK by plasma kallikrein has been implicated in the pathogenesis of inflammation and vascular injury. However, the functional role of HK in the absence of prekallikrein (PK), the proenzyme of plasma kallikrein, on vascular endothelial cells is not fully defined. In addition, no clinical abnormality is seen in PK-deficient patients. Therefore, an investigation into the effect of HK, in the absence of PK, on human pulmonary artery endothelial cell (HPAEC) function was performed. HK caused a marked and dose-dependent increase in the intracellular calcium [Ca(2+)](i) level in HPAEC. Gd(3+) and verapamil potentiated the HK-induced increase in [Ca(2+)](i). HK-induced Ca(2+) increase stimulated endothelial nitric oxide (NO) and prostacyclin (PGI(2)) production. The inhibitors of B(2) receptor-dependent signaling pathway impaired HK-mediated signal transduction in HPAEC. HK had no effect on endothelial permeability at physiological concentration. This study demonstrated that HK regulates endothelial cell function. HK could play an important role in maintaining normal endothelial function and blood flow and serve as a cardioprotective peptide.


Subject(s)
Cardiotonic Agents/pharmacology , Endothelial Cells/metabolism , Kininogen, High-Molecular-Weight/pharmacology , Receptor, Bradykinin B2/metabolism , Signal Transduction/drug effects , Calcium/metabolism , Cardiotonic Agents/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Endothelial Cells/cytology , Epoprostenol/biosynthesis , Gadolinium/pharmacology , Humans , Kininogen, High-Molecular-Weight/metabolism , Nitric Oxide/biosynthesis , Prekallikrein/metabolism , Pulmonary Artery/cytology , Pulmonary Artery/metabolism , Signal Transduction/physiology , Vasodilator Agents/pharmacology , Verapamil/pharmacology
6.
Am J Physiol Heart Circ Physiol ; 298(2): H652-8, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19966052

ABSTRACT

Inflammatory bowel disease and arthritis are associated with contact activation that results in cleavage of kininogen to form high molecular weight kininogen (HKa) and bradykinin. We have previously demonstrated that HKa can stimulate inflammatory cytokine and chemokine secretion from human monocytes. We now show that HKa can upregulate tissue factor antigen and procoagulant activity on human monocytes as a function of time (1-4 h) and HKa concentration (75-900 nM). The amino acid sequence responsible to block HKa effects is G440-H455. The HKa receptor macrophage-1 (Mac-1; CD11b18) is the binding site as shown by inhibition by a monoclonal antibody to CD11b/18. Chemical inhibitors of JNK, ERK, and p38 signaling pathways block cell signaling, as does an inhibitor to the transcription factor NF-kappaB. A combination of monoclonal antibodies to TNF-alpha and IL-1beta but neither alone inhibited the HKa induction of tissue factor. These results suggest that HKa mimics LPS by triggering a paracrine pathway in monocytes that depends on TNF-alpha and IL-1beta. Antibodies to kininogen or peptidomimetics might be a useful and safe therapy in inflammatory diseases or sepsis involving cytokines.


Subject(s)
Interleukin-1beta/metabolism , Kininogen, High-Molecular-Weight/pharmacology , Monocytes/metabolism , Thromboplastin/metabolism , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/drug effects , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Interleukin-1beta/immunology , Kininogen, High-Molecular-Weight/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Monocytes/cytology , Monocytes/drug effects , NF-kappa B/metabolism , RNA, Messenger/metabolism , Thromboplastin/genetics , Time Factors , Tumor Necrosis Factor-alpha/immunology , Up-Regulation/physiology
7.
Arterioscler Thromb Vasc Biol ; 29(10): 1602-7, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19661487

ABSTRACT

OBJECTIVE: Factor XI (FXI) promotes hemostasis and thrombosis through enhancement of thrombin generation and has been shown to play a critical role in the formation of occlusive thrombi in arterial injury models. The aim of this study was to investigate the mechanisms governing interactions between FXI and platelets. METHODS AND RESULTS: Platelet adhesion to immobilized FXI was abrogated in the presence of the low-density lipoprotein (LDL) receptor antagonist, receptor-associated protein (RAP), soluble recombinant apolipoprotein E receptor 2 (ApoER2), or the LDL-binding domain 1 or 2 of ApoER2. FXI supported wild-type murine platelet binding; in contrast, ApoER2-deficient murine platelets did not adhere to FXI. In the presence of shear, platelet aggregates formed on FXI or activated FXI (FXIa) surfaces, whereas the presence of RAP, binding domain 1 of ApoER2, or an anti-GPIb alpha mAb blocked platelet adhesion to FXI or FXIa under shear. Soluble FXI bound to immobilized ApoER2' with an affinity of 61 nmol/L. CONCLUSIONS: This study has identified apolipoprotein E receptor 2 (ApoER2, LRP8), a member of the LDL receptor family, as a platelet receptor for FXI. The interaction of FXI with other cell types that express ApoER2 remains to be explored.


Subject(s)
Factor XI/metabolism , Receptors, Lipoprotein/metabolism , Animals , Blood Platelets/metabolism , Calcium/metabolism , Humans , Kininogen, High-Molecular-Weight/pharmacology , LDL-Receptor Related Proteins , Ligands , Mice , Mice, Inbred C57BL , Platelet Adhesiveness , Zinc/pharmacology
8.
Oncogene ; 28(30): 2756-65, 2009 Jul 30.
Article in English | MEDLINE | ID: mdl-19483730

ABSTRACT

Upregulation and activation of epidermal growth factor receptor and/or urokinase-type plasminogen activator receptor in a variety of cancers have been shown to be associated with poor prognosis. High-molecular-weight kininogen can be hydrolysed by plasma kallikrein to bradykinin and cleaved high-molecular-weight kininogen (HKa). HKa and its domain 5 (D5) both have been shown to have potent anti-angiogenic activity. We now show that HKa blocks human prostate cancer cell (DU145) migration by 76.0+/-2.4% at 300 nM and invasion by 78.0+/-12.9% at 11.1 nM. D5 inhibits tumor migration and invasion in a concentration-dependent manner. Stimulation by basic fibroblast growth factor (bFGF) or vascular endothelial growth factor results in clustering of urokinase-type plasminogen activator receptor (uPAR) and epidermal growth factor receptor (EGFR) on the surface of DU145 cells. The co-localization of uPAR and EGFR is prevented by HKa. Immunoprecipitation suggests that uPAR, EGFR and alpha5beta1 integrin formed a ternary complex. Immunoblotting shows that HKa significantly decreases the bFGF-transactivated phosphorylation of EGFR at Tyr 1173 between 30 min and 4 h. The phosphorylation of extracellular signal-regulated kinase (ERK) and AKT, which are downstream effectors of EGFR, is also inhibited by HKa. These novel data indicate that HKa and D5 inhibit migration and invasion of human prostate cancer cells through an EGFR/uPAR pathway, suggesting the therapeutic potential of HKa and D5 to decrease metastasis of human prostate cancer.


Subject(s)
ErbB Receptors/physiology , Kininogen, High-Molecular-Weight/pharmacology , Prostatic Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Fibroblast Growth Factor 2/pharmacology , Humans , Kininogen, High-Molecular-Weight/chemistry , Kininogen, High-Molecular-Weight/therapeutic use , Male , Neoplasm Invasiveness , Prostatic Neoplasms/drug therapy , Protein Structure, Tertiary , Quinazolines , Receptors, Urokinase Plasminogen Activator/physiology , Signal Transduction/drug effects , Tyrphostins/pharmacology
9.
Proc Natl Acad Sci U S A ; 106(2): 570-5, 2009 Jan 13.
Article in English | MEDLINE | ID: mdl-19126685

ABSTRACT

Angiogenesis, the synthesis of new blood vessels from preexisting vessels, plays a critical role in normal wound healing and tumor growth. HKa (cleaved high molecular weight kininogen) is an endogenous inhibitor of angiogenesis formed by the cleavage of kininogen on endothelial cells. Ferritin is a protein principally known for its central role in iron storage. Here, we demonstrate that ferritin binds to HKa with high affinity (K(d) 13 nM). Further, ferritin antagonizes the antiangiogenic effects of HKa, enhancing the migration, assembly, and survival of HKa-treated endothelial cells. Effects of ferritin were independent of its iron content. Peptide mapping revealed that ferritin binds to a 22-aa subdomain of HKa that is critical to its antiangiogenic activity. In vivo, ferritin opposed HKa's antiangiogenic effects in a human prostate cancer xenograft, restoring tumor-dependent vessel growth. Ferritin-mediated regulation of angiogenesis represents a new angiogenic regulatory pathway, and identifies a new role for ferritin in cell biology.


Subject(s)
Ferritins/metabolism , Kininogen, High-Molecular-Weight/metabolism , Neovascularization, Pathologic , Cell Movement , Endothelial Cells/physiology , Ferritins/pharmacology , Humans , Kininogen, High-Molecular-Weight/pharmacology , Male , Prostatic Neoplasms/pathology , Protein Binding , Protein Interaction Mapping
10.
Exp Cell Res ; 314(4): 774-88, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18062965

ABSTRACT

Cleaved high molecular weight kininogen (HKa), as well as its domain 5 (D5), inhibits migration and proliferation induced by angiogenic factors and induces apoptosis in vitro. To study its effect on tube formation we utilized a collagen-fibrinogen, three-dimensional gel, an in vitro model of angiogenesis. HKa, GST-D5 and D5 had a similar inhibitory effect of tube length by 90+/-4.5%, 86+/-5.5% and 77+/-12.9%, respectively. D5-derived synthetic peptides: G440-H455 H475-H485 and G486-K502 inhibited tube length by 51+/-3.7%, 54+/-3.8% and 77+/-1.7%, respectively. By a comparison of its inhibitory potency and its sequences, a functional sequence of HKa was defined to G486-G496. PP2, a Src family kinase inhibitor, prevented tube formation in a dose-dependent manner (100-400 nM), but PP3 at 5 microM, an inactive analogue of PP2, did not. HKa and D5 inhibited Src 416 phosphorylation by 62+/-12.3% and 83+/-6.1%, respectively. The C-terminal Src kinase (Csk) inhibits Src kinase activity. Using a siRNA to Csk, expression of Csk was down-regulated by 86+/-7.0%, which significantly increased tube length by 27+/-5.8%. The addition of HKa and D5 completely blocked this effect. We further showed that HKa inhibited Src family kinase activity by disrupting the complex of uPAR, alphavbeta3 integrin and Src. Our results indicate that the anti-angiogenic effect of HKa and D5 is mediated at least in part through Src family kinases and identify a potential novel target for therapeutic inhibition of neovascularization in cancer and inflammatory arthritis.


Subject(s)
Endothelium, Vascular/cytology , Kininogen, High-Molecular-Weight/pharmacology , Neovascularization, Physiologic , src-Family Kinases/metabolism , Cell Adhesion , Cells, Cultured , Collagen/chemistry , Down-Regulation , Endothelium, Vascular/drug effects , Extracellular Matrix/chemistry , Fibrinogen/chemistry , Gels , Humans , Kininogen, High-Molecular-Weight/chemistry , Peptides/pharmacology , Protein Structure, Tertiary
11.
Am J Physiol Heart Circ Physiol ; 292(6): H2959-65, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17293494

ABSTRACT

High-molecular-weight kininogen (HK) and its domain 3 (D3) exhibit anticoagulant properties and inhibit platelet activation at low thrombin concentration in vitro. We hypothesized that the rapid occlusive thrombosis in HK-deficient (HKd) rats following endothelial injury of the aorta results from enhanced platelet aggregation by thrombin. The effects of D3 (G235-M357) or D3-derived peptides on thrombosis in vivo were tested. D3 and its exon 7C terminal peptide (E7CP, K270-Q292), expressed as glutathione S-transferase (GST) fusion proteins (GST-D3, GST-E7CP), or GST alone, as well as cleaved HK (HKa) or synthetic peptide E7CP, were infused intravenously 10 min before endothelial injury. Blood flow was reduced down to 10% of baseline flow within 28 +/- 5.2 min by a platelet-fibrin thrombus in GST-treated HKd rats compared with >240 min in GST-treated normal HK rats (wild type). GST-D3, GST-E7CP, HKa, or E7CP infusion prolonged the flow time to 233, >240, 223, and >240 min, respectively, in HKd rats. When GST-E7CP was infused 10 min after the injury, blood flow was maintained for >240 min. Thrombin-antithrombin concentrations were elevated by injury in HKd rats receiving GST from 35 to 55 microg/l and decreased with GST-E7CP, HKa, or E7CP reconstitution to 40, 15, and 9 microg/l, respectively. We conclude that HKd rats are prothrombotic and that HKa, kininogen D3, and its fragment E7CP modulate arterial thrombosis after endothelial injury.


Subject(s)
Aorta/metabolism , Endothelium, Vascular/metabolism , Fibrinolytic Agents/metabolism , Kininogen, High-Molecular-Weight/metabolism , Peptide Fragments/metabolism , Thrombosis/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified , Antithrombin III , Aorta/drug effects , Aorta/injuries , Aorta/pathology , Aorta/physiopathology , Blood Flow Velocity , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/injuries , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Fibrin/metabolism , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/therapeutic use , Glutathione Transferase/genetics , Kininogen, High-Molecular-Weight/chemistry , Kininogen, High-Molecular-Weight/genetics , Kininogen, High-Molecular-Weight/pharmacology , Kininogen, High-Molecular-Weight/therapeutic use , Male , Molecular Sequence Data , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Peptide Hydrolases/blood , Platelet Aggregation , Protein Structure, Tertiary , Rats , Rats, Inbred Lew/genetics , Recombinant Fusion Proteins/metabolism , Regional Blood Flow , Thrombin/metabolism , Thrombosis/pathology , Thrombosis/physiopathology , Thrombosis/prevention & control
12.
EMBO J ; 25(23): 5569-78, 2006 Nov 29.
Article in English | MEDLINE | ID: mdl-17093496

ABSTRACT

Activation of the contact system has two classical consequences: initiation of the intrinsic pathway of coagulation, and cleavage of high molecular weight kininogen (HK) leading to the release of bradykinin, a potent proinflammatory peptide. In human plasma, activation of the contact system at the surface of significant bacterial pathogens was found to result in further HK processing and bacterial killing. A fragment comprising the D3 domain of HK is generated, and within this fragment a sequence of 26 amino acids is mainly responsible for the antibacterial activity. A synthetic peptide covering this sequence kills several bacterial species, also at physiological salt concentration, as effectively as the classical human antibacterial peptide LL-37. Moreover, in an animal model of infection, inhibition of the contact system promotes bacterial dissemination and growth. These data identify a novel and important role for the contact system in the defence against invasive bacterial infection.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Immunity, Innate , Kininogen, High-Molecular-Weight/pharmacology , Peptides/pharmacology , Amino Acid Sequence , Animals , Anti-Bacterial Agents/chemistry , Humans , Kininogen, High-Molecular-Weight/chemistry , Kininogen, High-Molecular-Weight/metabolism , Mice , Molecular Sequence Data , Peptides/chemistry , Protein Structure, Tertiary
13.
J Biochem ; 140(6): 825-30, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17071946

ABSTRACT

High molecular weight (HMW) kininogen is known to be a large plasma protein and cleaved by plasma proteinase kallikrein, then it generates four fragments in the blood coagulation cascade: heavy chain, bradykinin, fragment 1.2, and light chain. The fragment 1.2 has also been found in the basic protein fraction of bovine milk as a bioactive protein which promotes osteoblast proliferation. The milk basic protein has been shown to be a multi functional edible protein which promotes bone formation and inhibits bone resorption. In the present study, we purified the fragment 1.2 from bovine plasma and assessed it could promote osteoblast proliferation and posses the activity after pepsin digestion. Purified plasma HMW kininogen did not promote the proliferation, however, the kallikrein-cleaved HMW kininogen promoted the proliferation. The fragment 1.2, purified from the proteolysate, also promoted the proliferation. The pepsin digestion was performed according to the method of the assessment of allergenesity of genetically modified crops. After pepsin digestion, the fragment 1.2 generated resistant fragments and showed the promoting activity of osteoblast proliferation. These results suggest that the enzymatically-digested fragments of bovine HMW kininogen are able to be a naturally occurred active protein that promotes the bone formation by oral administration.


Subject(s)
Kininogen, High-Molecular-Weight/pharmacology , Osteoblasts/cytology , Osteoblasts/drug effects , Peptide Fragments/pharmacology , 3T3 Cells , Animals , Bradykinin/pharmacology , Cattle , Cell Proliferation/drug effects , Kallikreins/metabolism , Mice , Osteogenesis/drug effects
14.
Arterioscler Thromb Vasc Biol ; 26(10): 2260-6, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16902163

ABSTRACT

OBJECTIVE: Plasma high-molecular-weight kininogen (HK) is cleaved in inflammatory diseases by kallikrein to HKa with release of bradykinin (BK). We postulated a direct link between HKa and cytokine/chemokine release. METHODS AND RESULTS: HKa, but not BK, releases cytokines tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta, IL-6, and chemokines IL-8 and MCP-1 from isolated human mononuclear cells. At a concentration of 600 nM, glutathione-S-transferase (GST) fusion proteins of kininogen domain 3 (D3), a fragment of domain 3, E7P (aaG255-Q292), HK domain 5 (D5), the D5 recombinant peptides HG (aa K420-D474) and HGK (aa H475-S626) stimulated secretion of IL-1beta from mononuclear cells. Monoclonal antibodies (MAbs) specific for D5 or specific for D3 blocked release of IL-1beta by HKa, supporting the importance of both domains. Antibodies to HK receptors on leukocytes including Mac-1, LFA-1, uPAR, and C1qR inhibited IL-1beta secretion induced by tKa 98%, 89%, 85%, and 62%, respectively. Fractionation of mononuclear cells identified the responsible cell, a blood monocyte. Inhibitors of signaling pathways NFkB, JNK, and p38 but not extracellular signal-regulated kinase (ERK) decreased cytokine release from mononuclear cells. HKa increased the synthesis of IL-1beta as deduced by an increase of IL-1beta mRNA at 1 to 2 hours. CONCLUSIONS: HKa domains 3 and 5 may contribute to the pathogenesis of inflammatory diseases by releasing IL-1beta from human monocytes using intracellular signaling pathways initiated by uPAR, beta2 integrins and gC1qR.


Subject(s)
Chemokines/metabolism , Cytokines/metabolism , Kininogen, High-Molecular-Weight/pharmacology , Macrophage-1 Antigen/metabolism , Membrane Glycoproteins/metabolism , Monocytes/metabolism , Receptors, Cell Surface/metabolism , Receptors, Complement/metabolism , Antibodies, Monoclonal/pharmacology , CD11a Antigen/immunology , Humans , Interleukin-1/antagonists & inhibitors , Interleukin-1/genetics , Interleukin-1/metabolism , Kininogen, High-Molecular-Weight/immunology , Kininogen, High-Molecular-Weight/metabolism , Macrophage-1 Antigen/immunology , Membrane Glycoproteins/immunology , Mitogen-Activated Protein Kinases/physiology , NF-kappa B/physiology , Osmolar Concentration , Peptide Fragments/immunology , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , RNA, Messenger/metabolism , Receptors, Cell Surface/immunology , Receptors, Complement/immunology , Receptors, Urokinase Plasminogen Activator , Time Factors
15.
Blood ; 107(12): 4714-20, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16418331

ABSTRACT

High-molecular-weight kininogen (HK) is an abundant plasma protein that plays a central role in activation of the kallikrein-kinin system. Cleavage of HK by plasma kallikrein results in release of the nonapeptide bradykinin (BK), leaving behind cleaved high-molecular-weight kininogen (HKa). Previous studies have demonstrated that HKa induces apoptosis of proliferating endothelial cells and inhibits angiogenesis in vivo, activities mediated primarily through its domain 5. However, the mechanisms by which these effects occur are not well understood. Here, we demonstrate that HKa induces apoptosis of endothelial cells cultured on gelatin, vitronectin, fibronectin, or laminin but not collagen type I or IV. The ability of HKa to induce endothelial-cell apoptosis is dependent on the generation of intracellular reactive oxygen species and associated with depletion of glutathione and peroxidation of endothelial-cell lipids, effects that occur only in cells cultured on matrix proteins permissive for HKa-induced apoptosis. Finally, the ability of HKa to induce endothelial-cell apoptosis is blocked by the addition of reduced glutathione or N-acetylcysteine. These studies demonstrate a unique role for oxidant stress in mediating the activity of an antiangiogenic polypeptide and highlight the importance of the extracellular matrix in regulating endothelial-cell survival.


Subject(s)
Apoptosis/physiology , Endothelial Cells/metabolism , Extracellular Matrix/metabolism , Kininogen, High-Molecular-Weight/metabolism , Neovascularization, Physiologic/physiology , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Apoptosis/drug effects , Bradykinin/metabolism , Cell Survival/drug effects , Cells, Cultured , Endothelial Cells/cytology , Extracellular Matrix Proteins/metabolism , Free Radical Scavengers/pharmacology , Glutathione/pharmacology , Humans , Kallikreins/metabolism , Kininogen, High-Molecular-Weight/pharmacology , Lipid Peroxidation/drug effects , Neovascularization, Physiologic/drug effects
16.
Thromb Haemost ; 94(3): 606-14, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16268479

ABSTRACT

Domain 5 (D5) of cleaved high molecular weight kininogen (HKa) inhibits angiogenesis in vivo and endothelial cell migration in vitro, but the cell signaling pathways involved in HKa and D5 inhibition of endothelial cell migration are incompletely delineated. This study examines the mechanism of HKa and D5 inhibition of two potent stimulators of endothelial cell migration, sphingosine 1-phosphate (S1P) and vascular endothelial growth factor (VEGF), that act through the P13-kinase-Akt signaling pathway. HKa and D5 inhibit bovine pulmonary artery endothelial cell (BPAE) or human umbilical vein endothelial cell chemotaxis in the modified-Boyden chamber in response toVEGF or S1P. The inhibition of migration by HKa is reversed by antibodies to urokinase-type plasminogen activator receptor. Both HKa and D5 decrease the speed of BPAE cell migration and alter the morphology in live, time-lapse microscopy after stimulation with S1P or VEGF. HKa and D5 reduce the localization of paxillin to the focal adhesions after S1P and VEGF stimulation. To better understand the intracellular signaling pathways, we examined the effect of HKa on the phosphorylation of Akt and its downstream effector, GSK-3alpha HKa and D5 inhibit phosphorylation of Akt and GSK-3alpha after stimulation withVEGF and S1P. Inhibitors of Akt and P13-kinase, the upstream activator of Akt, block endothelial cell migration and disrupt paxillin localization to the focal adhesions after stimulation with VEGF and S1P. Therefore we suggest that HKa through its D5 domain alters P13-kinase-Akt signaling to inhibit endothelial cell migration through alterations in the focal adhesions.


Subject(s)
Cell Movement/drug effects , Chemotaxis/drug effects , Endothelial Cells/drug effects , Kininogen, High-Molecular-Weight/pharmacology , Peptide Fragments/pharmacology , Signal Transduction/drug effects , Androstadienes/pharmacology , Animals , Cattle , Cells, Cultured , Chromones/pharmacology , Endothelial Cells/metabolism , Focal Adhesions/drug effects , Focal Adhesions/metabolism , Glycogen Synthase Kinase 3/metabolism , Humans , Kininogen, High-Molecular-Weight/chemistry , Lysophospholipids/pharmacology , Morpholines/pharmacology , Peptide Fragments/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Structure, Tertiary , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Time Factors , Vascular Endothelial Growth Factor A/pharmacology , Wortmannin
17.
Curr Cancer Drug Targets ; 5(7): 519-28, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16305348

ABSTRACT

High molecular weight kininogen (HK) is an abundant, multi-domain plasma protein that circulates in plasma primarily in its single chain form. Proteolytic cleavage of HK by plasma kallikrein releases the vasoactive nanopeptide bradykinin (BK), and converts HK into two-chain HK (HKa). BK appears to have pro-angiogenic activity, most likely mediated through binding to B1 and B2 receptors on endothelial cells. Conversely, HKa and its domain 5, but not (single chain) HK, have potent anti-angiogenic activity comparable to other endogenous angiogenesis inhibitors. The mechanism by which HKa exerts its anti-angiogenic activity remains controversial, but appears to involve binding to cell surface tropomyosin and induction of apoptosis of proliferating endothelial cells. A role for tropomyosin in mediating the anti-angiogenic signals of other anti-angiogenic proteins such as endostatin and histidine-proline-rich glycoprotein (HPRG) has also been reported. Here we review the physiological importance of high molecular weight kininogen in angiogenesis, with emphasis on the mechanism(s) by which this activity is mediated.


Subject(s)
Kininogen, High-Molecular-Weight/pharmacology , Neovascularization, Pathologic/prevention & control , Peptide Fragments/pharmacology , Amino Acid Sequence , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/pharmacology , Humans , Kininogen, High-Molecular-Weight/chemistry , Kininogen, High-Molecular-Weight/metabolism , Molecular Sequence Data , Plasma Kallikrein/metabolism
18.
Thromb Haemost ; 92(4): 811-9, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15467913

ABSTRACT

Evidence is accumulating to suggest that TFPI-2 is involved in regulating pericellular proteases implicated in a variety of physiologic and pathologic processes including cancer cell invasion, vascular inflammation, and atherosclerosis. Recent immunohistochemical studies of advanced atherosclerotic lesions, demonstrated a similar tissue distribution for TFPI-2, High Molecular Weight Kininogen (HK), and gC1qR/p33 (gC1qR), a ubiquitously expressed, multicompartmental cellular protein involved in modulating complement, coagulation, and kinin cascades. Further studies to evaluate TFPI-2 interactions with gC1qR demonstrated direct interactions between gC1qR and TFPI-2 using immunoprecipitation and solid phase binding studies. Specific and saturable binding between TFPI-2 and gC1qR (estimated Kd: approximately 70 nM) was observed by ELISA and surface plasmon resonance (Biacore) binding assays. Binding was inhibited by antibodies to gC1qR, and was strongly dependent on the Kunitz-2 domain of TFPI-2, as deletion of this domain reduced gC1qR-TFPI-2 interactions by approximately 75%. Deletion of gC1qR amino acids 74-95, involved in C1q binding, had no effect on gC1qR binding to TFPI-2, although antibodies to this region and purified C1q both inhibited binding, most likely via allosteric effects. In contrast, HK did not affect TFPI-2 binding to gC1qR. Binding of TFPI-2 to gC1qR produced statistically significant but modest reductions in TFPI-2 inhibition of plasmin, but had no effect on kallikrein inhibition in fluid phase chromogenic assays. Taken together, these data suggest that gC1qR may participate in tissue remodeling and inflammation by localizing TFPI-2 to the pericellular environment to modulate local protease activity and regulate HK activation.


Subject(s)
Endothelium, Vascular/pathology , Glycoproteins/metabolism , Hyaluronan Receptors/metabolism , Inflammation/etiology , Carrier Proteins , Complement C1q/pharmacology , Fibrinolysin/antagonists & inhibitors , Glycoproteins/pharmacology , Humans , Hyaluronan Receptors/pharmacology , Kallikreins/antagonists & inhibitors , Kininogen, High-Molecular-Weight/pharmacology , Mitochondrial Proteins , Protein Binding/drug effects
19.
J Biol Chem ; 279(44): 45470-6, 2004 Oct 29.
Article in English | MEDLINE | ID: mdl-15317813

ABSTRACT

Factor XI (FXI) is a homodimeric plasma zymogen that is cleaved at two internal Arg(369)-Ile(370) bonds by thrombin, factor XIIa, or factor XIa. FXI circulates as a complex with the glycoprotein high molecular weight kininogen (HK). FXI binds to specific sites (K(d) = approximately 10 nM, B(max) = approximately 1,500/platelet) on the surface of stimulated platelets, where it is efficiently activated by thrombin. The FXI Apple 3 (A3) domain mediates binding to platelets in the presence of HK and zinc ions (Zn(2+)) or prothrombin and calcium ions. The platelet glycoprotein (GP) Ib-IX-V complex is the receptor for FXI. Using surface plasmon resonance, we determined that FXI binds specifically to glycocalicin, the extracellular domain of GPIbalpha, in a Zn(2+)-dependent fashion (K(d) = approximately 52 nM). We now show that recombinant FXI A3 domain inhibits FXI inbinding to glycocalicin in the presence of Zn(2+), whereas the recombinant FXI A1, A2, or A4 domains have no effect. Experiments with full-length recombinant FXI mutants show that, in the presence of Zn(2+), glycocalicin binds FXI at a heparin-binding site in A3 (Lys(252) and Lys(253)) and not by amino acids previously shown to be required for platelet binding (Ser(248), Arg(250), Lys(255), Phe(260), and Gln(263)). However, binding in the presence of HK and Zn(2+) requires Ser(248), Arg(250), Lys(255), Phe(260), and GLn(263) and not Lys(252) and Lys(253). Thus, binding of FXI to GPIbalpha is mediated by amino acids in the A3 domain in the presence or absence of HK. This interaction is important for the initiation of the consolidation phase of blood coagulation and the generation of thrombin at sites of platelet thrombus formation.


Subject(s)
Factor XI/chemistry , Membrane Proteins/metabolism , Binding Sites , Blood Platelets/metabolism , Chlorides/pharmacology , Factor XI/metabolism , Humans , Kininogen, High-Molecular-Weight/pharmacology , Membrane Glycoproteins , Platelet Glycoprotein GPIb-IX Complex/metabolism , Zinc Compounds/pharmacology
20.
Circ Res ; 94(9): 1227-34, 2004 May 14.
Article in English | MEDLINE | ID: mdl-15044324

ABSTRACT

Cleaved high molecular weight kininogen (HKa) has been shown to inhibit in vivo neovascularization and induce apoptosis of endothelial cells. We have shown that HKa-induced apoptosis correlated with its antiadhesive effect and was regulated by extracellular matrix (ECM) proteins. In this study, we identified the urokinase-type plasminogen activator receptor (uPAR) as a target of HKa activity at the endothelial cell surface. Anti-uPAR antibodies blocked the apoptotic effect of HKa. Further studies revealed that uPAR formed a signaling complex containing integrin alpha(v)beta3 or alpha5beta1, caveolin, and Src kinase Yes in endothelial cells. HKa physically disrupted the formation of this complex in a manner that paralleled its apoptotic effect. For the first time, our results provide a mechanistic explanation for the previous observation that HKa selectively induces apoptosis of endothelial cells grown on vitronectin, but not cells grown on fibronectin. These data also resolve the controversial role of uPAR in mediating the apoptotic and antiadhesive activities of HKa.


Subject(s)
Apoptosis/drug effects , Kininogen, High-Molecular-Weight/physiology , Receptors, Cell Surface/physiology , src-Family Kinases , Antibodies, Monoclonal/pharmacology , Apoptosis/physiology , Caveolin 1 , Caveolins/physiology , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cells, Cultured , Endothelial Cells , Endothelium, Vascular , Extracellular Matrix Proteins/physiology , Fibroblast Growth Factor 2/pharmacology , Fibronectins/pharmacology , Humans , Integrin alpha5beta1/physiology , Integrin alphaVbeta3/physiology , Kininogen, High-Molecular-Weight/pharmacology , Macromolecular Substances , Phosphatidylinositol Diacylglycerol-Lyase/pharmacology , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-yes , Receptors, Cell Surface/drug effects , Receptors, Urokinase Plasminogen Activator , Umbilical Veins , Vitronectin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...