Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Expert Opin Biol Ther ; 19(12): 1233-1245, 2019 12.
Article in English | MEDLINE | ID: mdl-31657963

ABSTRACT

Introduction: Hereditary angioedema due to C1 inhibitor deficiency (C1-INH-HAE) is a rare yet still probably underdiagnosed clinical condition. Recurrent episodes of subcutaneous and sub-mucosal swelling may involve the skin, the gastrointestinal tract or even the upper airways, exposing the patients to the risk of death. With the aim of improving patients' quality of life, the therapeutic scenario has expanded over the years.Areas covered: The focus of the present review is lanadelumab, a fully human, κ-light-chain, monoclonal immunoglobulin G1 against plasma kallikrein, currently approved for long-term prophylaxis of C1-INH-HAE attacks in the USA and Canada and designated as an orphan drug by the European Medicines Agency.Expert opinion: Lanadelumab is able to inhibit plasma kallikrein with high selectivity and affinity. The subsequent phases of drug development and the ongoing open-label trial have proven its safety and efficacy. It overcomes some of the limitations of other drugs available for long-term prophylaxis, given the easy route of administration, the simple administration schedule and the possibility to tailor the treatment to each patient. Further studies are needed to test its efficacy also in other types of angioedema for which a central role of plasma kallikrein is envisaged.


Subject(s)
Angioedemas, Hereditary/drug therapy , Antibodies, Monoclonal, Humanized/therapeutic use , Immunoglobulin G/therapeutic use , Plasma Kallikrein/immunology , Antibodies, Monoclonal, Humanized/immunology , Humans , Immunoglobulin G/immunology , Plasma Kallikrein/antagonists & inhibitors , Quality of Life
2.
Biochem Biophys Res Commun ; 518(3): 409-415, 2019 10 20.
Article in English | MEDLINE | ID: mdl-31451224

ABSTRACT

Particulate matter (PM) is a key component of air pollutants and is associated with mortality of cardiovascular and respiratory diseases. PM-induced tissue injury involves inflammation and coagulation. Plasma prekallikrein (pKal), along with coagulation factor XII (FXII) and high-molecular-weight kininogen (HK), form the plasma kallikrein-kinin system (KKS), a component of the innate immune response that generates proinflammatory products in response to injury. When the KKS proteins contact with activation surface such as negatively charged molecules, this system becomes activated. Activated kallikrein (Kal) activates FXII to initiate the intrinsic coagulation pathway, and cleaves HK to release bradykinin to enhance vascular permeability and systemic inflammation. In his study we determined the role of plasma pKal in the PM2.5-induced lung injury. Using TALEN technology, we generated a new mouse strain lacking the gene for pKal. In PM2.5-induced lung injury model, Klkb1-/- mice exhibited a decrease in total protein, cells numbers in bronchoalveolar lavage fluid (BALF) and histologic lung injury score. The TNF-α and IL-6 levels in BALF were significantly decreased in PM2.5-treated Klkb1-/- mice. Plasma thrombin-antithrombin (TAT) complex levels were significantly decreased in PM2.5-treated Klkb1-/- mice. PM2.5 induces pKal activation, HK cleavage and bradykinin production. PM2.5-induced HK cleavage in plasma was completely blocked by a Kal inhibitor, as well as in pKal-deficient plasma. PM2.5 markedly induced thrombin generation in human plasma and wild-type mouse plasma, which was inhibited by both blockade and deficiency of pKal. Taken together, plasma pKal is activated by PM2.5 and the activated Kal plays an important role in PM2.5-induced lung injury.


Subject(s)
Blood Coagulation , Inflammation/etiology , Lung Injury/etiology , Particulate Matter/adverse effects , Plasma Kallikrein/immunology , Animals , Gene Deletion , Humans , Inflammation/blood , Inflammation/genetics , Inflammation/immunology , Lung Injury/blood , Lung Injury/genetics , Lung Injury/immunology , Mice , Mice, Knockout , Particulate Matter/immunology , Plasma Kallikrein/analysis , Plasma Kallikrein/genetics
3.
Int J Parasitol ; 49(5): 337-346, 2019 04.
Article in English | MEDLINE | ID: mdl-30796952

ABSTRACT

Serine peptidases are involved in many physiological processes including digestion, haemostasis and complement cascade. Parasites regulate activities of host serine peptidases to their own benefit, employing various inhibitors, many of which belong to the Kunitz-type protein family. In this study, we confirmed the presence of potential anticoagulants in protein extracts of the haematophagous monogenean Eudiplozoon nipponicum which parasitizes the common carp. We then focused on a Kunitz protein (EnKT1) discovered in the E. nipponicum transcriptome, which structurally resembles textilinin-1, an antihemorrhagic snake venom factor from Pseudonaja textilis. The protein was recombinantly expressed, purified and biochemically characterised. The recombinant EnKT1 did inhibit in vitro activity of Factor Xa of the coagulation cascade, but exhibited a higher activity against plasmin and plasma kallikrein, which participate in fibrinolysis, production of kinins, and complement activation. Anti-coagulation properties of EnKT1 based on the inhibition of Factor Xa were confirmed by thromboelastography, but no effect on fibrinolysis was observed. Moreover, we discovered that EnKT1 significantly impairs the function of fish complement, possibly by inhibiting plasmin or Factor Xa which can act as a C3 and C5 convertase. We localised Enkt1 transcripts and protein within haematin digestive cells of the parasite by RNA in situ hybridisation and immunohistochemistry, respectively. Based on these results, we suggest that the secretory Kunitz protein of E. nipponicum has a dual function. In particular, it impairs both haemostasis and complement activation in vitro, and thus might facilitate digestion of a host's blood and protect a parasite's gastrodermis from damage by the complement. This study presents, to our knowledge, the first characterisation of a Kunitz protein from monogeneans and the first example of a parasite Kunitz inhibitor that impairs the function of the complement.


Subject(s)
Complement System Proteins/immunology , Fish Diseases/immunology , Helminth Proteins/immunology , Hemostasis , Trematoda/immunology , Trematode Infections/veterinary , Amino Acid Sequence , Animals , Anticoagulants/chemistry , Anticoagulants/immunology , Antifibrinolytic Agents/chemistry , Antifibrinolytic Agents/immunology , Carps/blood , Carps/immunology , Carps/parasitology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/immunology , Factor Xa/immunology , Factor Xa Inhibitors/chemistry , Factor Xa Inhibitors/immunology , Fibrinolysin/immunology , Fish Diseases/blood , Fish Diseases/parasitology , Helminth Proteins/chemistry , Helminth Proteins/genetics , Host-Parasite Interactions , Plasma Kallikrein/antagonists & inhibitors , Plasma Kallikrein/immunology , Sequence Alignment , Trematoda/chemistry , Trematoda/genetics , Trematode Infections/blood , Trematode Infections/immunology , Trematode Infections/parasitology
4.
Scand J Immunol ; 72(2): 128-33, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20618771

ABSTRACT

Some components of the kinin system such as plasma kallikrein levels, the activities of tissue kallikrein (including saliva) and kininase II and the concentrations of kininogen fractions (low-molecular weight/LKg and high-molecular weight/HKg) were evaluated in the plasma of patients with thromboangiitis obliterans (TAO) presenting clinical symptoms of the condition. Twenty TAO were diagnosed by means of the traditional Shionoya and Olin criteria and later classified into non-smokers (n = 11) and active smokers (n = 9). Fifty-three normal, non-smoking/smoking individuals (control) were also studied. Kininogen levels were determined by ELISA; the activities of kallikreins and kininase II were determined using selective substrates. The levels of enzymes (kallikreins and kininase II) and protein (kininogens) were significantly higher in patients with TAO who were active smokers compared to the control groups (no matter whether control individuals were active smokers or non-smokers, P < 0.001 for all comparisons). Interestingly, regardless of the time of disease onset, a significant increase in the levels of these components of the kinin system was also observed in patients when TAO active smokers were compared with TAO ex-smokers (P < 0.01 for all analysed parameters). Activation of the kinin system in patients with TAO may indicate the involvement of vasodilatation in an attempt to control vascular changes, thereby favouring the deposition of immune complexes at the vascular level because of nicotine stimulation. Moreover, our results corroborate the idea that TAO can be an autoimmune disorder with specific mechanisms.


Subject(s)
Kininogens/immunology , Peptidyl-Dipeptidase A/immunology , Plasma Kallikrein/immunology , Thromboangiitis Obliterans/immunology , Tissue Kallikreins/immunology , Adult , Female , Humans , Kininogens/blood , Male , Middle Aged , Peptidyl-Dipeptidase A/blood , Plasma Kallikrein/analysis , Smoking/blood , Smoking/immunology , Statistics, Nonparametric , Thromboangiitis Obliterans/enzymology , Tissue Kallikreins/analysis
5.
Allergy Asthma Proc ; 30(5): 487-92, 2009.
Article in English | MEDLINE | ID: mdl-19843402

ABSTRACT

Hereditary angioedema (HAE) was first described in the 19th century. Over the past 50 years, many details of the pathophysiology and molecular biology of HAE have been elucidated. Two types of HAE, type I and type II, result from mutations in the gene for the broad-spectrum protease inhibitor C1 inhibitor (C1INH). Type I HAE is characterized by low antigenic and functional C1INH levels and type II HAE has normal antigenic but low functional C1INH levels. Type III HAE, by contrast, has normal antigenic and functional C1INH levels. In some families, type III HAE has been linked to mutations in Hageman factor. C1INH is the primary inhibitor of the complement proteases C1r and C1s as well as the contact system proteases activated Hageman factor (coagulation factor XIIa and XIIf) and plasma kallikrein. It is also an inhibitor of plasmin and coagulation factor XIa. The primary mediator of swelling in HAE has now been unequivocally shown to be bradykinin, generated from activation of the plasma contact system. The knowledge gained concerning the underlying mechanisms of the different types of HAE allow the clinician to approach the laboratory diagnosis with confidence and provides opportunities for novel therapeutic strategies.


Subject(s)
Angioedemas, Hereditary/diagnosis , Complement C1 Inactivator Proteins/metabolism , Factor XII/metabolism , Angioedemas, Hereditary/genetics , Angioedemas, Hereditary/immunology , Bradykinin/immunology , Bradykinin/metabolism , Capillary Permeability/immunology , Capillary Permeability/physiology , Complement C1 Inactivator Proteins/genetics , Complement C1 Inactivator Proteins/immunology , Complement C1 Inhibitor Protein , Complement C1r/immunology , Complement C1r/metabolism , Complement C1s/immunology , Complement C1s/metabolism , Complement C2/immunology , Complement C2/metabolism , Factor XII/immunology , Humans , Mannose-Binding Protein-Associated Serine Proteases/immunology , Mannose-Binding Protein-Associated Serine Proteases/metabolism , Plasma Kallikrein/immunology , Plasma Kallikrein/metabolism
6.
Allergy Asthma Proc ; 30(5): 500-5, 2009.
Article in English | MEDLINE | ID: mdl-19843404

ABSTRACT

Remarkable progress has been made in understanding the molecular mechanisms underlying attacks of swelling in hereditary angioedema (HAE). Treatment options in the United States for this potentially life-threatening disease had remained essentially static, however, over the past 40 years. Prophylactic therapy had relied on attenuated androgens or antifibrinolytic agents. Although demonstrably effective, these drugs have been fraught with side effects. Acute therapy has been largely relegated to supportive care. In this article we discuss emerging treatments that have evolved from the recognition that kinin generation is the fundamental abnormality leading to attacks of angioedema. We will review the newly approved replacement therapy for prophylaxis of HAE attacks with C1 inhibitor (C1INH). Potential options for the acute treatment of HAE will be discussed including purified C1INH, recombinant C1INH, an inhibitor of plasma kallikrein, and a B2-receptor antagonist. The arrival of these novel therapies promises to transform the future management of HAE.


Subject(s)
Angioedemas, Hereditary/drug therapy , Bradykinin/analogs & derivatives , Complement C1 Inhibitor Protein/therapeutic use , Enzyme Replacement Therapy , Plasma Kallikrein/metabolism , Receptors, Bradykinin/metabolism , Recombinant Proteins/therapeutic use , Bradykinin/immunology , Bradykinin/metabolism , Bradykinin/therapeutic use , Bradykinin Receptor Antagonists , Capillary Permeability/drug effects , Capillary Permeability/immunology , Clinical Trials as Topic , Complement C1 Inhibitor Protein/administration & dosage , Humans , Plasma Kallikrein/antagonists & inhibitors , Plasma Kallikrein/immunology , Receptors, Bradykinin/immunology , Recombinant Proteins/administration & dosage
7.
Scand J Clin Lab Invest ; 63(6): 425-9, 2003.
Article in English | MEDLINE | ID: mdl-14594323

ABSTRACT

This study presents immunological and functional evidence to suggest that the activation of prekallikrein (PK) in human plasma yields two modifications of kallikrein. One of these modifications showed amidolytic properties strongly deviating from those registered for the main part of the enzyme. The substrates were S-2302, Bz-Pro-Phe-Arg-pNA, S-2366 and S-2222. In PAGE immunoblots the PK heavy chain mAb 13G11 was found to detect the kallikrein 85 kD double band and bands with mol. weights of about 152 and 135 kD. Such a 152 kD band could be removed together with an IgG fraction on a Protein G column. In this study the kallikrein identity was confirmed by an estimation of the levels obtained in amidolytic assays of mixtures of normal plasma and plasma deficient in FXI, which in immunological assays showed a PK level of 140-150%, of normal. A comparison in amidolytic assays of normal plasma and plasma from patients with Crohn's disease showed that patients' plasma contained a significantly higher level than normal of modified kallikrein. An IgG removal procedure removed all modified kallikrein, and did not affect ordinary kallikrein levels.


Subject(s)
Crohn Disease/blood , Immunoglobulin G/immunology , Plasma Kallikrein/immunology , Amidohydrolases/blood , Antibody Affinity , Crohn Disease/immunology , Electrophoresis, Polyacrylamide Gel , Factor XI/analysis , Factor XII/analysis , Humans , Immunoassay , Plasma Kallikrein/analysis , Plasma Kallikrein/metabolism , Prekallikrein/metabolism
8.
Int Immunopharmacol ; 2(13-14): 1867-73, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12489801

ABSTRACT

Plasma prekallikrein (PPK), the zymogen of the contact phase protease plasma kallikrein, forms a non-covalent complex with its substrate H-kininogen (HK). HK binds to cell surface proteoglycans, indirectly anchoring this bradykinin-generating protease to endothelial cells. The heavy chain of PPK consisting of four apple domains designated A1 to A4. Previous studies indicated that a major HK binding site on PPK is within the A2 domain, with additional contributions to binding provided by the N-terminal portion of Al and the central part of A4. To precisely map the relevant binding segments in A2, we employed a monoclonal anti-PPK antibody (PKH6) that binds to A2 and blocks HK-PPK complex formation with an apparent IC50 of 8 nM. Using recombinant A2 C-terminal deletion mutants, we mapped the target epitope of PKH6 to the N-terminal portion of A2, residues 92-153. C-terminal deletion of A2 to residue 145 resulted in a loss of PKH6 binding, as did proteolytic cleavage of A2 at Lys140-Arg141. A comparison of HK binding to various A2 deletion mutants revealed that the major HK binding site is localized to residues 145-153 in the central portion of A2, where it overlaps with the PKH6 epitope. This sequence is conserved in the A2 domain of the related protease factor XI, explaining the unusual strong cross-reactivity of PHK6 with factor XI, as well as the similar HK-binding characteristics of PPK and factor XI.


Subject(s)
Epitopes/metabolism , Kininogen, High-Molecular-Weight/metabolism , Plasma Kallikrein/metabolism , Prekallikrein/metabolism , Antibodies, Monoclonal/immunology , Binding Sites , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Epitope Mapping , Epitopes/immunology , Humans , Immunoassay , Plasma Kallikrein/genetics , Plasma Kallikrein/immunology , Prekallikrein/genetics , Prekallikrein/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Sequence Deletion
9.
Metab Brain Dis ; 15(4): 315-23, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11383556

ABSTRACT

Plasma kallikrein (PK) is a cofactor in blood coagulation and modulates inflammation through the release of bradykinin. Previously it was believed that plasma prekallikrein (PPK), the precursor of PK and a member of the serine protease superfamily, was synthesized exclusively by hepatocytes and secreted into circulation. However, recent studies show that the human brain contains a high level of PPK mRNA. In this study we sought to determine which areas of the brain express PK. Tissue from the spinal cord and 13 different regions of the human brain were collected at autopsy within 24h from death. Sections were probed using polyclonal antibodies (characterized by Western blotting) specific for PK. PK concentrations in extracts of these tissues were measured by ELISA. Immunolabeling of PK was observed in the cell bodies of the neurons of the hypothalamus, thalamus, spinal cord, cerebral cortex and brainstem. Positive PK immuno-reactivity was also demonstrated in the cytoplasm of the ependymal cells in sections of the hypothalamus and spinal cord. In addition, some fibre tracts of the pons, medulla and hippocampus as well as secretory cells of the pituitary gland also labeled. No immunoreactive PK was visualized in the choroid plexus or cerebellum. Our data demonstrate the cellular localization of PK in human brain. This work is supported by other studies that demonstrate PK mRNA in human heart, lung, trachea and brain. The cellular distribution of PK and kinin receptors in specific brain areas suggests a role for PK in the nervous system.


Subject(s)
Brain Chemistry , Plasma Kallikrein/analysis , Adolescent , Adult , Aged , Antibodies , Child , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Male , Middle Aged , Neurons/chemistry , Plasma Kallikrein/immunology , Spinal Cord/chemistry , Spinal Cord/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...