Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Biol Macromol ; 266(Pt 2): 131065, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38521329

RESUMO

Protein C inhibitor (PCI) maintains hemostasis by inhibiting both procoagulant and anticoagulant serine proteases, and plays important roles in coagulation, fibrinolysis, reproduction, and anti-angiogenesis. The reactive site loop of PCI traps and irreversibly inhibits the proteases like APC (activating protein C), thrombin (FIIa) and factor Xa (FXa). Previous studies on antithrombin (ATIII) had identified Tyr253 and Glu255 as functional exosites that interact and aid in the inhibition of factor IXa and FXa. Presence of exosite in PCI is not known, however a sequence comparison with the PCI from different vertebrate species and ATIII identified Glu239 to be absolutely conserved. PCI residues analogous to ATIII exosite residues were mutated to R238A and E239A. Purified variant PCI in the presence of heparin (10 µg/ml) showed a 2-4 fold decrease in the rate of inhibition of the proteases. However, the stoichiometry of inhibition of FIIa, APC, and FXa by native PCI, R238A and E239A variants were found to be close to 1.0, which also indicated the formation of stable complexes based on SDS-PAGE and western blot analysis with thrombin and APC. Our findings revealed the possible presence of an exosite in PCI that influences the protease inhibition rates.


Assuntos
Heparina , Inibidor da Proteína C , Serina Proteases , Inibidor da Proteína C/química , Inibidor da Proteína C/metabolismo , Heparina/química , Heparina/farmacologia , Humanos , Serina Proteases/metabolismo , Serina Proteases/química , Trombina/metabolismo , Proteína C/metabolismo , Proteína C/química , Fator Xa/metabolismo , Fator Xa/química , Sequência de Aminoácidos , Ativação Enzimática/efeitos dos fármacos
2.
Exp Cell Res ; 391(1): 111987, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32240661

RESUMO

BACKGROUND: The protein plasminogen activator inhibitor-1 (PAI-1), an inhibitor specific for urokinase plasminogen activator (uPA) and tissue plasminogen activator (tPA), has been shown to have a key role in cancer metastases. Currently, it is unknown as to whether the exocellular inhibition of PAI-1 can inhibit the migration of cancer cells. METHODS: By fusing the mutated serine protease domain (SPD) of uPA and human serum albumin (HSA), PAItrap3, a protein that traps PAI-1, was synthesized and experiments were conducted to determine if exocellular PAItrap3 attenuates PAI-1-induced cancer cell migration in vitro. RESULTS: PAItrap3 (0.8 µM) significantly inhibited the motility of MCF-7, MDA-MB-231, HeLa and 4T1 cancer cells, by 90%, 50%, 30% and 20%, respectively, without significantly altering their proliferation. The PAI-1-induced rearrangement of F-actin was significantly inhibited by PAItrap3, which produced a decrease in the number of cell protrusions by at least 20%. CONCLUSIONS: In vitro, PAItrap3 inhibited PAI-1-induced cancer cell migration, mainly through inhibiting the rearrangement of F-actin. Overall, these results, provided they can be extrapolated to humans, suggest that the PAItrap3 protein could be used as an exocellular inhibitor to attenuate cancer metastases.


Assuntos
Actinas/genética , Movimento Celular/efeitos dos fármacos , Inibidor 1 de Ativador de Plasminogênio/farmacologia , Inibidor da Proteína C/farmacologia , Actinas/antagonistas & inibidores , Actinas/metabolismo , Sítios de Ligação , Linhagem Celular , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Células HeLa , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Histidina/genética , Histidina/metabolismo , Humanos , Células MCF-7 , Oligopeptídeos/genética , Oligopeptídeos/metabolismo , Pichia/genética , Pichia/metabolismo , Inibidor 1 de Ativador de Plasminogênio/química , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Ligação Proteica , Inibidor da Proteína C/química , Inibidor da Proteína C/genética , Inibidor da Proteína C/metabolismo , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
3.
Adv Exp Med Biol ; 966: 93-101, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28639251

RESUMO

It is generally accepted that the phospholipid bilayer of the cell membrane is impermeable for proteins and peptides and that these molecules require special mechanisms for their transport from the extra- to the intracellular space. Recently there is increasing evidence that certain proteins/peptides can also directly cross the phospholipid membrane. SERPINA5 (protein C inhibitor) is a secreted protease inhibitor with broad protease reactivity and wide tissue distribution. It binds glycosaminoglycans and certain phospoholipids, which can modulate its inhibitory activity. SERPINA5 has been shown to be internalized by platelets, granulocytes, HL-60 promyelocytic leukemia cells, and by Jurkat lymphoma cells. Once inside the cell it can translocate to the nucleus. There are several indications that SERPINA5 can directly cross the phospholipid bilayer of the cell membrane. In this review we will describe what is known so far about the conditions, as well as the cellular and molecular requirements for SERPINA5 translocation through the cell membrane and for its penetration of pure phospholipid vesicles.


Assuntos
Permeabilidade da Membrana Celular , Membrana Celular/metabolismo , Inibidor da Proteína C/metabolismo , Animais , Humanos , Inibidor da Proteína C/química , Conformação Proteica , Transporte Proteico , Relação Estrutura-Atividade
4.
Arch Biochem Biophys ; 604: 128-42, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27372899

RESUMO

Pro-coagulant, anti-coagulant and fibrinolytic pathways are responsible for maintaining hemostatic balance under physiological conditions. Any deviation from these pathways would result in hypercoagulability leading to life threatening diseases like myocardial infarction, stroke, portal vein thrombosis, deep vein thrombosis (DVT) and pulmonary embolism (PE). Angiogenesis is the process of sprouting of new blood vessels from pre-existing ones and plays a critical role in vascular repair, diabetic retinopathy, chronic inflammation and cancer progression. Serpins; a superfamily of protease inhibitors, play a key role in regulating both angiogenesis and coagulation. They are characterized by the presence of highly conserved secondary structure comprising of 3 ß-sheets and 7-9 α-helices. Inhibitory role of serpins is modulated by binding to cofactors, specially heparin and heparan sulfate proteoglycans (HSPGs) present on cell surfaces and extracellular matrix. Heparin and HSPGs are the mainstay of anti-coagulant therapy and also have therapeutic potential as anti-angiogenic inhibitors. Many of the heparin binding serpins that regulate coagulation cascade are also potent inhibitors of angiogenesis. Understanding the molecular mechanism of the switch between their specific anti-coagulant and anti-angiogenic role during inflammation, stress and regular hemostasis is important. In this review, we have tried to integrate the role of different serpins, their interaction with cofactors and their interplay in regulating coagulation and angiogenesis.


Assuntos
Coagulação Sanguínea , Heparina/química , Neovascularização Patológica , Serpinas/fisiologia , Angiotensinogênio/química , Animais , Anticoagulantes/química , Antitrombinas/química , Proteínas Sanguíneas/química , Vasos Sanguíneos/fisiopatologia , Progressão da Doença , Matriz Extracelular/química , Proteínas do Olho/química , Fibrinólise , Proteoglicanas de Heparan Sulfato/química , Heparina de Baixo Peso Molecular/química , Homeostase , Humanos , Inflamação , Conformação Molecular , Fatores de Crescimento Neural/química , Inibidor 1 de Ativador de Plasminogênio/química , Inibidor da Proteína C/química , Serpina E2/química , Serpinas/química , Transdução de Sinais
5.
PLoS One ; 10(11): e0143137, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26580551

RESUMO

Protein C inhibitor is a secreted, non-specific serine protease inhibitor with broad protease reactivity. It binds glycosaminoglycans and anionic phospholipids, which can modulate its activity. Anionic phospholipids, such as phosphatidylserine are normally localized to the inner leaflet of the plasma membrane, but are exposed on activated and apoptotic cells and on plasma membrane-derived microparticles. In this report we show by flow cytometry that microparticles derived from cultured cells and activated platelets incorporated protein C inhibitor during membrane blebbing. Moreover, protein C inhibitor is present in/on microparticles circulating in normal human plasma as judged from Western blots, ELISAs, flow cytometry, and mass spectrometry. These plasma microparticles are mainly derived from megakaryocytes. They seem to be saturated with protein C inhibitor, since they do not bind added fluorescence-labeled protein C inhibitor. Heparin partially removed microparticle-bound protein C inhibitor, supporting our assumption that protein C inhibitor is bound via phospholipids. To assess the biological role of microparticle-bound protein C inhibitor we performed protease inhibition assays and co-precipitated putative binding partners on microparticles with anti-protein C inhibitor IgG. As judged from amidolytic assays microparticle-bound protein C inhibitor did not inhibit activated protein C or thrombin, nor did microparticles modulate the activity of exogenous protein C inhibitor. Among the proteins co-precipitating with protein C inhibitor, complement factors, especially complement factor 3, were most striking. Taken together, our data do not support a major role of microparticle-associated protein C inhibitor in coagulation, but rather suggest an interaction with proteins of the complement system present on these phospholipid vesicles.


Assuntos
Plaquetas/química , Membrana Celular/química , Micropartículas Derivadas de Células/química , Megacariócitos/química , Inibidor da Proteína C/química , Proteína C/antagonistas & inibidores , Adulto , Plaquetas/citologia , Membrana Celular/metabolismo , Micropartículas Derivadas de Células/metabolismo , Feminino , Heparina/química , Humanos , Imunoglobulina G/química , Imunoglobulina G/metabolismo , Células Jurkat , Masculino , Megacariócitos/citologia , Pessoa de Meia-Idade , Fosfolipídeos/química , Fosfolipídeos/metabolismo , Fator Plaquetário 3/química , Fator Plaquetário 3/metabolismo , Ligação Proteica , Proteína C/metabolismo , Inibidor da Proteína C/metabolismo , Trombina/química , Trombina/metabolismo
6.
Thromb Res ; 135(6): 1203-8, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25887633

RESUMO

INTRODUCTION: Protein C inhibitor (PCI), a member of the serine protease inhibitor family, is expressed in various human tissues, including liver and kidneys. In the plasma, PCI physiologically inhibits an anticoagulant serine protease, activated protein C (APC). PCI expressed by cancer cells suppresses tumor invasion by inhibiting urokinase-type plasminogen activator, and inhibits tumor growth and metastasis, which are independent of its protease-inhibitory activity. In the present study, we clarified the effects of host PCI on growth and metastasis of B16 melanoma (B16) cells by comparing between wild-type mice and mice transgenic for human PCI gene (hPCI-TG), which have a tissue distribution of PCI similar to that observed in humans. MATERIALS AND METHODS: Growth of intracutaneously-injected B16 cells was evaluated by measuring the tumor volume, and metastatic behavior of intravenously-injected B16 cells by counting the number of metastatic lung nodules. RESULTS: Growth of intracutaneously injected B16 cells was significantly faster in wild-type mice than in hPCI-TG mice; however, hPCI-TG mice developed more metastatic nodules of B16 cells in the lungs. Immunohistochemical analysis using anti-mouse fibrinogen antibody revealed more fibrin deposition in the lung in hPCI-TG mice than in wild-type mice. Furthermore, the more invasive behavior observed in hPCI-TG mice was reduced by rabbit anti-human PCI IgG, APC, or soluble TM administration for 3 consecutive days including the day that B16 cells were injected. CONCLUSIONS: Our results suggest that like PCI expressed in tumor cells, host PCI also inhibits tumor growth, but host PCI promotes tumor metastasis via its procoagulant properties.


Assuntos
Coagulantes/química , Neoplasias Pulmonares/tratamento farmacológico , Inibidor da Proteína C/sangue , Proteína C/antagonistas & inibidores , Trombofilia/sangue , Animais , Feminino , Humanos , Técnicas Imunoenzimáticas , Imuno-Histoquímica , Masculino , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Metástase Neoplásica , Transplante de Neoplasias , Inibidor da Proteína C/química , RNA/análise , Trombomodulina/química , Fatores de Tempo
7.
PLoS One ; 10(4): e0122410, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25830552

RESUMO

Ischaemic stroke is caused by occlusive thrombi in the cerebral vasculature. Although tissue-plasminogen activator (tPA) can be administered as thrombolytic therapy, it has major limitations, which include disruption of the blood-brain barrier and an increased risk of bleeding. Treatments that prevent or limit such deleterious effects could be of major clinical importance. Activated protein C (APC) is a natural anticoagulant that regulates thrombin generation, but also confers endothelial cytoprotective effects and improved endothelial barrier function mediated through its cell signalling properties. In murine models of stroke, although APC can limit the deleterious effects of tPA due to its cell signalling function, its anticoagulant actions can further elevate the risk of bleeding. Thus, APC variants such as APC(5A), APC(Ca-ins) and APC(36-39) with reduced anticoagulant, but normal signalling function may have therapeutic benefit. Human and murine protein C (5A), (Ca-ins) and (36-39) variants were expressed and characterised. All protein C variants were secreted normally, but 5-20% of the protein C (Ca-ins) variants were secreted as disulphide-linked dimers. Thrombin generation assays suggested reductions in anticoagulant function of 50- to 57-fold for APC(36-39), 22- to 27-fold for APC(Ca-ins) and 14- to 17-fold for APC(5A). Interestingly, whereas human wt APC, APC(36-39) and APC(Ca-ins) were inhibited similarly by protein C inhibitor (t½ - 33 to 39 mins), APC(5A) was inactivated ~9-fold faster (t½ - 4 mins). Using the murine middle cerebral artery occlusion ischaemia/repurfusion injury model, in combination with tPA, APC(36-39), which cannot be enhanced by its cofactor protein S, significantly improved neurological scores, reduced cerebral infarct area by ~50% and reduced oedema ratio. APC(36-39) also significantly reduced bleeding in the brain induced by administration of tPA, whereas wt APC did not. If our data can be extrapolated to clinical settings, then APC(36-39) could represent a feasible adjunctive therapy for ischaemic stroke.


Assuntos
Anticoagulantes/uso terapêutico , Infarto da Artéria Cerebral Média/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Proteína C/uso terapêutico , Animais , Anticoagulantes/farmacologia , Avaliação Pré-Clínica de Medicamentos , Células HEK293 , Humanos , Infarto da Artéria Cerebral Média/sangue , Cinética , Masculino , Camundongos Endogâmicos C57BL , Mutação de Sentido Incorreto , Fármacos Neuroprotetores/farmacologia , Proteína C/química , Proteína C/farmacologia , Inibidor da Proteína C/química , Inibidor da Proteína C/farmacologia , Proteólise , Traumatismo por Reperfusão/sangue , Traumatismo por Reperfusão/prevenção & controle , Trombina/metabolismo , Tempo de Trombina
8.
J Biomol Struct Dyn ; 33(1): 85-92, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-24251463

RESUMO

Activated Protein C (APC) is a multifunctional serine protease, primarily known for its anticoagulant function in the coagulation system. Several studies have already elucidated its role in counteracting apoptosis and inflammation in cells, while significant effort is still ongoing for defining its involvement in sepsis. Earlier literature has shown that the antiseptic function of APC is mediated by its binding to leukocyte integrins, which is due to the presence of the integrin binding motif Arg-Gly-Asp at the N-terminus of the APC catalytic chain. Many natural mutants have been identified in patients with Protein C deficiency diagnosis including a variant of specificity pocket (Gly216Asp). In this work, we present a molecular model of the complex of APC with αVß3 integrin obtained by protein-protein docking approach. A computational analysis of this variant is hereby presented, based on molecular dynamics and docking simulations, aiming at investigating the effects of the Gly216Asp mutation on the protein conformation and inferring its functional implications. Our study shows that such mutation is likely to impair the protease activity while preserving the overall protein fold. Moreover, superposition of the integrin binding motifs in wild-type and mutant forms suggests that the interaction with integrin can still occur and thus the mutant is likely to retain its antiseptic function related to the neutrophyl integrin binding. Therapeutic applications could result in this APC mutant which retains antiseptic function without anticoagulant side effects.


Assuntos
Integrina alfaVbeta3/química , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Peptídeo Hidrolases/química , Proteína C/química , Anti-Infecciosos Locais/química , Anti-Infecciosos Locais/metabolismo , Sítios de Ligação/genética , Humanos , Integrina alfaVbeta3/metabolismo , Mutação de Sentido Incorreto , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/metabolismo , Ligação Proteica , Proteína C/genética , Proteína C/metabolismo , Inibidor da Proteína C/química , Inibidor da Proteína C/metabolismo , Conformação Proteica , Estrutura Terciária de Proteína
9.
J Biol Chem ; 290(5): 3081-91, 2015 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-25488662

RESUMO

Protein C inhibitor (PCI) is a serpin with broad protease reactivity. It binds glycosaminoglycans and certain phospholipids that can modulate its inhibitory activity. PCI can penetrate through cellular membranes via binding to phosphatidylethanolamine. The exact mechanism of PCI internalization and the intracellular role of the serpin are not well understood. Here we showed that testisin, a glycosylphosphatidylinositol-anchored serine protease, cleaved human PCI and mouse PCI (mPCI) at their reactive sites as well as at sites close to their N terminus. This cleavage was observed not only with testisin in solution but also with cell membrane-anchored testisin on U937 cells. The cleavage close to the N terminus released peptides rich in basic amino acids. Synthetic peptides corresponding to the released peptides of human PCI (His(1)-Arg(11)) and mPCI (Arg(1)-Ala(18)) functioned as cell-penetrating peptides. Because intact mPCI but not testisin-cleaved mPCI was internalized by Jurkat T cells, a truncated mPCI mimicking testisin-cleaved mPCI was created. The truncated mPCI lacking 18 amino acids at the N terminus was not taken up by Jurkat T cells. Therefore our model suggests that testisin or other proteases could regulate the internalization of PCI by removing its N terminus. This may represent one of the mechanisms regulating the intracellular functions of PCI.


Assuntos
Peptídeos Penetradores de Células/química , Peptídeos Penetradores de Células/metabolismo , Inibidor da Proteína C/química , Inibidor da Proteína C/metabolismo , Animais , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular/fisiologia , Proteínas Ligadas por GPI/metabolismo , Humanos , Camundongos , Serina Endopeptidases/metabolismo , Células U937
10.
Virology ; 458-459: 11-21, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24928035

RESUMO

HIV-exposed seronegative individuals (HESNs) are persons who remain seronegative despite repeated exposure to HIV, suggesting an in vivo resistance mechanism to HIV. Elucidation of endogenous factors responsible for this phenomenon may aid in the development of new classes of microbicides and therapeutics. We compared cervicovaginal protein abundance profiles between high-risk HESN and two control groups: low-risk HESN and HIV-positives. Four iTRAQ-based quantitative experiments were performed using samples classified based on presence/absence of particular gynaecological conditions. After statistical analysis, two proteins were shown to be differentially abundant between high-risk HESNs and control groups. Serpin A5, a serine proteinase inhibitor and Myeloblastin, a serine protease, were up- and downregulated, respectively. Commercially available ELISA assays were used to confirm differential Serpin A5 levels. These results suggest that HIV resistance in CVF of HESNs is the result of a delicate balance between two complementary mechanisms: downregulation of serine proteinases and upregulation of their inhibitors.


Assuntos
Líquidos Corporais/enzimologia , Infecções por HIV/imunologia , HIV-1/fisiologia , Inibidor da Proteína C/metabolismo , Serina Proteases/metabolismo , Côte d'Ivoire/epidemiologia , Feminino , Regulação Enzimológica da Expressão Gênica/imunologia , Predisposição Genética para Doença , Infecções por HIV/epidemiologia , Infecções por HIV/virologia , Humanos , Inibidor da Proteína C/química , Inibidor da Proteína C/genética , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Serina Proteases/genética , Profissionais do Sexo
11.
Thromb Res ; 133(6): 1105-14, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24513148

RESUMO

INTRODUCTION: Activated protein C (APC) is the central enzyme of the anticoagulant protein C pathway. Low concentrations of APC circulate in plasma and are believed to contribute to the maintenance of a normal haemostatic balance. MATERIALS AND METHODS: We have used a structure-based virtual screening approach to discover small drug-like molecules that inhibit the interaction between APC and its substrate FVa through inhibition of a predominant APC exosite, known to be involved in FVa substrate binding. We have combined in silico selection with functional screening and direct binding analysis to identify novel molecules and to ascertain and characterize the inhibition of the interaction between APC and FVa. RESULTS: We have identified a number of novel molecules that bind to APC and protein C with Kd values in the range of 10(-3)- 10(-5)M. Inhibition by these molecules is incomplete, which most likely reflects the extended surface that is involved in the interaction between APC and its substrates. Direct binding of hit molecules to variant APC molecules that were mutated in the targeted binding site revealed that several of the molecules presented a 100-500 fold lower affinity for the variant molecule, suggesting that these molecules indeed bind the exosite of APC. CONCLUSIONS: The protein-protein interaction inhibitors discovered here, could function as starting molecules for further development of small molecules with anti-APC properties. Such molecules may be of clinical interest, in particular in individuals where thrombin formation is compromised and the haemostatic balance is tipped towards bleeding tendencies, such as in haemophilia A.


Assuntos
Inibidor da Proteína C/farmacologia , Proteína C/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/farmacologia , Sítios de Ligação , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Modelos Moleculares , Proteína C/química , Proteína C/metabolismo , Inibidor da Proteína C/química , Bibliotecas de Moléculas Pequenas/química , Relação Estrutura-Atividade , Ressonância de Plasmônio de Superfície
12.
Bioorg Med Chem Lett ; 24(3): 821-7, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24418773

RESUMO

A design strategy was used to identify inhibitors of activated protein C with selectivity over thrombin featured by a basic and/or aromatic functionality for binding to the S2 pocket. Our strongest inhibitor showed an IC50-material value and selectivity for APC vs thrombin similar to a compound previously reported in the literature. However, in contrast to the reference compound, our compound showed a retained coagulant effect of thrombin with increasing substrate concentration in a modified Calibrated Automated Thrombogram (CAT) method. This was likely related to our compound being inactive against FVIIa, while the reference compound showed an IC50 of 8.9 µM. Thus, the higher selectivity of our compound against all relevant coagulation factors likely explained its higher therapeutic potential in comparison to the reference compound. The data indicate that at least a 100-fold selectivity over other serine proteases in the coagulation cascade will be required for an effective APC inhibitor.


Assuntos
Desenho de Fármacos , Inibidor da Proteína C/síntese química , Inibidor da Proteína C/farmacologia , Trombina/antagonistas & inibidores , Sítios de Ligação , Coagulantes/síntese química , Coagulantes/química , Coagulantes/farmacologia , Hemofilia A/tratamento farmacológico , Concentração Inibidora 50 , Ligação Proteica/efeitos dos fármacos , Inibidor da Proteína C/química , Relação Estrutura-Atividade , Especificidade por Substrato
13.
Thromb Haemost ; 111(1): 41-52, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24136361

RESUMO

Protein C inhibitor (PCI, SerpinA5) is a heparin-binding serpin which can penetrate through cellular membranes. Selected negatively charged phospholipids like unsaturated phosphatidylserine and oxidised phosphatidylethanolamine bind to PCI and stimulate its inhibitory activity towards different proteases. The interaction of phospholipids with PCI might also alter the lipid distribution pattern of blood cells and influence the remodelling of cellular membranes. Here we showed that PCI is an additional binding partner of phosphatidic acid (PA), cardiolipin (CL), and phosphoinositides (PIPs). Protein lipid overlay assays exhibited a unique binding pattern of PCI towards different lipid species. In addition PA, CL, and unsaturated, monophosphorylated PIPs stimulated the inhibitory property of PCI towards activated protein C in a heparin like manner. As shown for kallistatin (SerpinA4) and vaspin (SerpinA12), the incubation of cells with PCI led to the activation of protein kinase B (AKT), which could be achieved through direct interaction of PCI with PIPs. This model is supported by the fact that PCI stimulated the PIP-dependent 5-phosphatase SHIP2 in vitro, which would result in AKT activation. Hence the interaction of PCI with different lipids might not only stimulate the inhibition of potential target protease by PCI, but could also alter intracellular lipid signalling.


Assuntos
Lipídeos/química , Monoéster Fosfórico Hidrolases/química , Inibidor da Proteína C/química , Proteína C/antagonistas & inibidores , Coagulação Sanguínea , Cardiolipinas/química , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Diglicerídeos/química , Relação Dose-Resposta a Droga , Ácidos Graxos/química , Células HEK293 , Heparina/química , Humanos , Ácidos Fosfatídicos/química , Fosfatidiletanolaminas/química , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases , Fosfatidilserinas/química , Fosfolipídeos/química , Ligação Proteica , Proteínas Recombinantes/química , Serpinas/química , Transdução de Sinais
14.
Bioarchitecture ; 2(4): 118-23, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22960705

RESUMO

The PCI fold is based on a stack of α-helices topped with a winged-helix domain and is found in a range of proteins that form central parts of large complexes such as the proteasome lid, the COP9 signalosome, elongation factor eIF3, and the TREX-2 complex. Recent structural determinations have given intriguing insight into how these folds function both to facilitate the generation of larger proteinaceous assembles and also to interact functionally with nucleic acids.


Assuntos
Inibidor da Proteína C/química , Inibidor da Proteína C/metabolismo , Dobramento de Proteína , Humanos , Modelos Moleculares , Ácidos Nucleicos/química , Ácidos Nucleicos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Estrutura Secundária de Proteína , Proteínas de Saccharomyces cerevisiae/metabolismo
15.
J Proteome Res ; 11(3): 1868-78, 2012 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-22257466

RESUMO

We have investigated the precision of peptide quantitation by MALDI-TOF mass spectrometry (MS) using six pairs of proteotypic peptides (light) and same-sequence stable isotope labeled synthetic internal standards (heavy). These were combined in two types of dilution curves spanning 100-fold and 2000-fold ratios. Coefficients of variation (CV; standard deviation divided by mean value) were examined across replicate MALDI spots using a reflector acquisition method requiring 100 000 counts for the most intense peak in each summed spectrum. The CV of light/heavy peptide centroid peak area ratios determined on four replicate spots per sample, averaged across 11 points of a 100-fold dilution curve and over all six peptides, was 2.2% (ranging from 1.5 to 3.7% among peptides) at 55 fmol total (light + heavy) of each peptide applied per spot, and 2.5% at 11 fmol applied. The average CV of measurements at near-equivalence (light = heavy, the center of the dilution curve) for the six peptides was 1.0%, about 17-fold lower CV than that observed when five peptides were ratioed to a sixth peptide (i.e., a different-sequence internal standard). Response curves across the 100-fold range were not completely linear but could be closely modeled by a power law fit giving R(2) values >0.998 for all peptides. The MALDI-TOF MS method was used to determine the endogenous level of a proteotypic peptide (EDQYHYLLDR) of human protein C inhibitor (PCI) in a plasma digest after enrichment by capture on a high affinity antipeptide antibody, a technique called stable isotope standards and capture by anti-peptide antibodies (SISCAPA). The level of PCI was determined to be 770 ng/mL with a replicate measurement CV of 1.5% and a >14 000-fold target enrichment via SISCAPA-MALDI-TOF. These results indicate that MALDI-TOF technology can provide precise quantitation of high-to-medium abundance peptide biomarkers over a 100-fold dynamic range when ratioed to same-sequence labeled internal standards and enriched to near purity by specific antibody capture. The robustness and throughput of MALDI-TOF in comparison to conventional nano-LC-MS technology could enable currently impractical large-scale verification studies of protein biomarkers.


Assuntos
Fragmentos de Peptídeos/química , Sequência de Aminoácidos , Calibragem , Humanos , Dados de Sequência Molecular , Peso Molecular , Fragmentos de Peptídeos/sangue , Inibidor da Proteína C/sangue , Inibidor da Proteína C/química , Proteólise , Proteômica , Padrões de Referência , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/normas , Tripsina/química
16.
Thromb Res ; 130(4): 661-6, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22206708

RESUMO

Protein C inhibitor was purified from human plasma by use of a dermatan sulfate or heparin column, followed by hydroxyapatite, gel filtration and ion exchange columns. A dimer of protein C inhibitor was detected by SDS-PAGE under reducing conditions, in addition to two forms of monomer species. One of the monomers, 52-kDa PCI, formed a stable complex with activated protein C, urokinase, plasma and tissue kallikrein, but the dimer species and 48-kDa PCI were inactive. When the monomer and dimer forms of protein C inhibitor were applied to 2D-PAGE, more than 20 spots were observed by Western blot analysis and were confirmed to be protein C inhibitor by MALDI-TOF mass spectrometry. The heterogeneity of the protein C inhibitor species was not due to glycosylation or phosphorylation.


Assuntos
Inibidor da Proteína C/química , Inibidor da Proteína C/metabolismo , Sequência de Aminoácidos , Dermatan Sulfato/química , Eletroforese em Gel de Poliacrilamida , Humanos , Dados de Sequência Molecular , Calicreína Plasmática/metabolismo , Proteína C/antagonistas & inibidores , Proteína C/metabolismo , Inibidor da Proteína C/isolamento & purificação , Multimerização Proteica , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Calicreínas Teciduais/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
17.
PLoS One ; 6(12): e29011, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22205989

RESUMO

Protein C inhibitor (PCI) is a serpin type of serine protease inhibitor that is found in many tissues and fluids in human, including blood plasma, seminal plasma and urine. This inhibitor displays an unusually broad protease specificity compared with other serpins. Previous studies have shown that the N-glycan(s) and the NH2-terminus affect some blood-related functions of PCI. In this study, we have for the first time determined the N-glycan profile of seminal plasma PCI, by mass spectrometry. The N-glycan structures differed markedly compared with those of both blood-derived and urinary PCI, providing evidence that the N-glycans of PCI are expressed in a tissue-specific manner. The most abundant structure (m/z 2592.9) had a composition of Fuc3Hex5HexNAc4, consistent with a core fucosylated bi-antennary glycan with terminal Lewis(x). A major serine protease in semen, prostate specific antigen (PSA), was used to evaluate the effects of N-glycans and the NH2-terminus on a PCI function related to the reproductive tract. Second-order rate constants for PSA inhibition by PCI were 4.3±0.2 and 4.1±0.5 M⁻¹ s⁻¹ for the natural full-length PCI and a form lacking six amino acids at the NH2-terminus, respectively, whereas these constants were 4.8±0.1 and 29±7 M⁻¹ s⁻¹ for the corresponding PNGase F-treated forms. The 7-8-fold higher rate constants obtained when both the N-glycans and the NH2-terminus had been removed suggest that these structures jointly affect the rate of PSA inhibition, presumably by together hindering conformational changes of PCI required to bind to the catalytic pocket of PSA.


Assuntos
Regulação da Expressão Gênica , Polissacarídeos , Inibidor da Proteína C/química , Inibidor da Proteína C/metabolismo , Humanos , Masculino , Modelos Moleculares , Especificidade de Órgãos , Antígeno Prostático Específico/antagonistas & inibidores , Antígeno Prostático Específico/química , Antígeno Prostático Específico/metabolismo , Inibidor da Proteína C/sangue , Inibidor da Proteína C/farmacologia , Conformação Proteica , Sêmen/metabolismo
18.
Methods Enzymol ; 501: 105-37, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22078533

RESUMO

Serpins (serine protease inhibitors) have traditionally been grouped together based on structural homology. They share common structural features of primary sequence, but not all serpins require binding to cofactors in order to achieve maximal protease inhibition. In order to obtain physiologically relevant rates of inhibition of target proteases, some serpins utilize the unbranched sulfated polysaccharide chains known as glycosaminoglycans (GAGs) to enhance inhibition. These GAG-binding serpins include antithrombin (AT), heparin cofactor II (HCII), and protein C inhibitor (PCI). The GAGs heparin and heparan sulfate have been shown to bind AT, HCII, and PCI, while HCII is also able to utilize dermatan sulfate as a cofactor. Other serpins such as PAI-1, kallistatin, and α(1)-antitrypsin also interact with GAGs with different endpoints, some accelerating protease inhibition while others inhibit it. There are many serpins that bind or carry ligands that are unrelated to GAGs, which are described elsewhere in this work. For most GAG-binding serpins, binding of the GAG occurs in a conserved region of the serpin near or involving helix D, with the exception of PCI, which utilizes helix H. The binding of GAG to serpin can lead to a conformational change within the serpin, which can lead to increased or tighter binding to the protease, and can accelerate the rates of inhibition up to 10,000-fold compared to the unbound native serpin. In this chapter, we will discuss three major GAG-binding serpins with known physiological roles in modulating coagulation: AT (SERPINC1), HCII (SERPIND1), and PCI (SERPINA5). We will review methodologies implemented to study the structure of these serpins and those used to study their interactions with GAG's. We discuss novel techniques to examine the serpin-GAG interaction and finally we review the biological roles of these serpins by describing the mouse models used to study them.


Assuntos
Antitrombina III/metabolismo , Técnicas de Química Analítica , Cofator II da Heparina/metabolismo , Biologia Molecular/métodos , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Inibidor da Proteína C/metabolismo , alfa 1-Antitripsina/metabolismo , Animais , Antitrombina III/química , Sítios de Ligação , Coagulação Sanguínea , Dermatan Sulfato/química , Dermatan Sulfato/metabolismo , Heparina/química , Heparina/metabolismo , Cofator II da Heparina/química , Humanos , Cinética , Camundongos , Camundongos Knockout , Modelos Moleculares , Inibidor 1 de Ativador de Plasminogênio/química , Ligação Proteica , Inibidor da Proteína C/química , Estrutura Secundária de Proteína , Especificidade da Espécie , Trombina/química , Trombina/metabolismo , alfa 1-Antitripsina/química
19.
Semin Thromb Hemost ; 37(4): 349-54, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21805440

RESUMO

Protein C inhibitor (PCI) is a serine protease inhibitor and was originally identified as an inhibitor of activated protein C (APC). However, PCI is not specific for APC and also inhibits several proteases involved in coagulation, fibrinolysis, cancer, wound healing, and fertility. The biological function of PCI is unknown due to broad enzyme specificity, its wide tissue distribution, and the lack of a suitable animal model. This review highlights the specific roles of PCI in the areas of hemostasis and thrombosis and fertilization, and it also describes the latest information on the fascinating participation of the protein in intracellular processes, phospholipid binding, and killing of bacteria.


Assuntos
Inibidor da Proteína C/fisiologia , Animais , Fibrinólise/fisiologia , Hemostasia/fisiologia , Humanos , Proteína C/antagonistas & inibidores , Inibidor da Proteína C/sangue , Inibidor da Proteína C/química , Trombose/metabolismo
20.
Biochem Biophys Res Commun ; 403(2): 198-202, 2010 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-21056543

RESUMO

Protein C inhibitor (PCI) is a 57-kDa glycoprotein that exists in many tissues and secretions in human. As a member of the serpin superfamily of proteins it displays unusually broad protease specificity. PCI is implicated in the regulation of a wide range of processes, including blood coagulation, fertilization, prevention of tumors and pathogen defence. It has been reported that PCI isolated from human blood plasma is highly heterogeneous, and that this heterogeneity is caused by differences in N-glycan structures, N-glycosylation occupancy, and the presence of two forms that differ by the presence or absence of 6 amino acids at the amino-terminus. In this study we have verified that such heterogeneity exists in PCI purified from single individuals, and that individuals of two different ethnicities possess a similar PCI pattern, verifying that the micro-heterogeneity is conserved among humans. Furthermore, we have provided experimental evidence that PCI in both individuals is O-glycosylated on Thr20 with a core type 1 O-glycan, which is mostly NeuAcGalGalNAc. Modeling suggested that the O-glycan attachment site is located in proximity to several ligand-binding sites of the inhibitor.


Assuntos
Polissacarídeos/química , Inibidor da Proteína C/química , Sequência de Aminoácidos , Doadores de Sangue , Glicosilação , Humanos , Dados de Sequência Molecular , Conformação Proteica , Espectrometria de Massas por Ionização por Electrospray
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...