Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Clin Transl Sci ; 13(6): 1208-1216, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32407574

RESUMO

Hereditary angioedema (HAE) with C1 inhibitor deficiency is a rare disorder characterized by unpredictable, potentially life-threatening recurrent angioedema attacks. Lanadelumab is a fully human monoclonal antibody with selective binding to active plasma kallikrein, and prevents the formation of cleaved high molecular weight kininogen (cHMWK) and bradykinin, thereby preventing HAE attacks. The clinical pharmacology of lanadelumab was characterized following subcutaneous administration in 257 subjects (24 healthy subjects and 233 patients with HAE). The pharmacokinetics of lanadelumab were described using a one-compartment model with first-order rate of absorption and linear clearance, showing slow absorption and a long half-life (14.8 days). A covariate analysis retained body weight and health status on apparent clearance (CL/F) and body weight on volume of distribution (V/F). Population estimates of CL/F and V/F were 0.0249 L/hour (0.586 L/day) and 12.8 L, respectively. An indirect-response Imax model showed 53.7% maximum suppression in cHMWK formation with a low potential for interactions with concomitant medications (analgesic, anti-inflammatory, and antirheumatic medications). A 300 mg dose administered Q2W was associated with a mean steady-state minimum concentration (Cmin,ss ; 25.4 µg/mL) that was ~ 4.5-fold higher than the half-maximal inhibitory concentration for cHMWK reduction (5.71 µg/mL). Exposure-response analyses suggest that 300 mg Q2W dosing was associated with a significantly reduced HAE attack rate, prolonged time to first attack after treatment initiation, and lower need for concomitant medications. The response was comparable across patient body weight groups. Findings from this analysis support the dosing rationale for lanadelumab to prevent attacks in patients with HAE.


Assuntos
Angioedemas Hereditários/tratamento farmacológico , Anticorpos Monoclonais Humanizados/farmacocinética , Calicreína Plasmática/antagonistas & inibidores , Prevenção Secundária/métodos , Adolescente , Adulto , Idoso , Angioedemas Hereditários/sangue , Anticorpos Monoclonais Humanizados/administração & dosagem , Área Sob a Curva , Bradicinina/metabolismo , Criança , Conjuntos de Dados como Assunto , Relação Dose-Resposta a Droga , Interações Medicamentosas , Feminino , Voluntários Saudáveis , Humanos , Injeções Subcutâneas , Cininogênios/metabolismo , Masculino , Pessoa de Meia-Idade , Calicreína Plasmática/metabolismo , Ensaios Clínicos Controlados Aleatórios como Assunto , Resultado do Tratamento , Adulto Jovem
2.
J Agric Food Chem ; 68(9): 2673-2683, 2020 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-32050765

RESUMO

Nonalcoholic fatty liver disease (NAFLD) is rapidly becoming the leading cause of chronic liver diseases throughout the world. The deficit of pharmacotherapy for NAFLD calls for an urgent need for a new drug discovery and lifestyle management. Black tea is the most popular and functional drink consumed worldwide. Its main bioactive constituent theaflavin helps to prevent obesity-a major risk factor for NAFLD. To find new targets for the development of effective and safe therapeutic drugs from natural plants for NAFLD, we found a theaflavin monomer theaflavin-3,3'-digallate (TF3), which significantly reduced lipid droplet accumulation in hepatocytes, and directly bound and inhibited the activation of plasma kallikrein (PK), which was further proved to stimulate adenosine monophosphate activated protein kinase (AMPK) and its downstream targets. Taken together, we proposed that the TF3-PK-AMPK regulatory axis is a novel mechanism of lipid deposition mitigation, and PK could be a new target for NAFLD treatment.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Biflavonoides/farmacologia , Catequina/farmacologia , Hepatócitos/metabolismo , Gotículas Lipídicas/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Extratos Vegetais/farmacologia , Calicreína Plasmática/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Apoptose/efeitos dos fármacos , Camellia sinensis/química , Células Hep G2 , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Humanos , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/fisiopatologia , Transdução de Sinais/efeitos dos fármacos
3.
Expert Opin Investig Drugs ; 29(3): 237-244, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31985300

RESUMO

Introduction: Plasma kallikrein is a  mediator of vascular leakage and inflammation. Activation of plasma kallikrein can induce features of diabetic macular edema (DME) in preclinical models. Human vitreous shows elevated plasma kallikrein levels in patients with DME. Because of the incomplete response of some patients to anti-VEGF agents, and the treatment burden associated with frequent dosing, there is still considerable need for VEGF-independent targeted pathways.Areas covered: This review covers the role of plasma kallikrein in the pathogenesis of DME and the therapeutic potential of plasma kallikrein inhibitors. It discusses early clinical studies of plasma kallikrein pathway modulation for DME, which have been associated with some improvement in visual acuity but with limited improvement in macular edema. This review also highlights KVD001, which is furthest along the development pathway, THR-149, which has recently completed a phase 1 study, and oral agents under development.Expert opinion: Plasma kallikrein inhibitors have a potential role in the treatment of DME, with mixed functional/anatomic results in early clinical trials. Given the large unmet need in DME treatment, further studies are warranted.


Assuntos
Retinopatia Diabética/tratamento farmacológico , Edema Macular/tratamento farmacológico , Calicreína Plasmática/antagonistas & inibidores , Animais , Retinopatia Diabética/fisiopatologia , Desenvolvimento de Medicamentos , Drogas em Investigação/farmacologia , Humanos , Edema Macular/fisiopatologia , Calicreína Plasmática/metabolismo
4.
Arterioscler Thromb Vasc Biol ; 40(1): 103-111, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31766871

RESUMO

OBJECTIVES: FXIa (factor XIa) induces clot formation, and human congenital FXI deficiency protects against venous thromboembolism and stroke. In contrast, the role of FXI in hemostasis is rather small, especially compared with FIX deficiency. Little is known about the cause of the difference in phenotypes associated with FIX deficiency and FXI deficiency. We speculated that activation of FIX via the intrinsic coagulation is not solely dependent on FXI(a; activated FXI) and aimed at identifying an FXI-independent FIX activation pathway. Approach and Results: We observed that ellagic acid and long-chain polyphosphates activated the coagulation system in FXI-deficient plasma, as could be demonstrated by measurement of thrombin generation, FIXa-AT (antithrombin), and FXa-AT complex levels, suggesting an FXI bypass route of FIX activation. Addition of a specific PKa (plasma kallikrein) inhibitor to FXI-deficient plasma decreased thrombin generation, prolonged activated partial thromboplastin time, and diminished FIXa-AT and FXa-AT complex formation, indicating that PKa plays a role in the FXI bypass route of FIX activation. In addition, FIXa-AT complex formation was significantly increased in F11-/- mice treated with ellagic acid or long-chain polyphosphates compared with controls and this increase was significantly reduced by inhibition of PKa. CONCLUSIONS: We demonstrated that activation of FXII leads to thrombin generation via FIX activation by PKa in the absence of FXI. These findings may, in part, explain the different phenotypes associated with FIX and FXI deficiencies.


Assuntos
Coagulação Sanguínea/fisiologia , Fator IX/metabolismo , Deficiência do Fator XI/sangue , Fator XI/metabolismo , Calicreína Plasmática/metabolismo , Trombina/metabolismo , Trombose/sangue , Animais , Modelos Animais de Doenças , Deficiência do Fator XI/complicações , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Trombose/etiologia
5.
J Struct Biol ; 206(2): 170-182, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30876891

RESUMO

Plasma kallikrein (pKal) is a serine protease responsible for cleaving high-molecular-weight kininogen to produce the pro-inflammatory peptide, bradykinin. Unregulated pKal activity can lead to hereditary angioedema (HAE) following excess bradykinin release. HAE attacks can lead to a compromised airway that can be life threatening. As there are limited agents for prophylaxis of HAE attacks, there is a high unmet need for a therapeutic agent for regulating pKal with a high degree of specificity. Here we present crystal structures of both full-length and the protease domain of pKal, bound to two very distinct classes of small-molecule inhibitors: compound 1, and BCX4161. Both inhibitors demonstrate low nM inhibitory potency for pKal and varying specificity for related serine proteases. Compound 1 utilizes a surprising mode of interaction and upon binding results in a rearrangement of the binding pocket. Co-crystal structures of pKal describes why this class of small-molecule inhibitor is potent. Lack of conservation in surrounding residues explains the ∼10,000-fold specificity over structurally similar proteases, as shown by in vitro protease inhibition data. Structural information, combined with biochemical and enzymatic analyses, provides a novel scaffold for the design of targeted oral small molecule inhibitors of pKal for treatment of HAE and other diseases resulting from unregulated plasma kallikrein activity.


Assuntos
Calicreína Plasmática/química , Células Endoteliais da Veia Umbilical Humana , Humanos , Cininogênio de Alto Peso Molecular/metabolismo , Cininogênios/metabolismo , Calicreína Plasmática/antagonistas & inibidores , Calicreína Plasmática/metabolismo , Ligação Proteica , Conformação Proteica , Bibliotecas de Moléculas Pequenas/farmacologia
6.
Platelets ; 30(3): 305-313, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-29442535

RESUMO

The purpose of antithrombotic therapy is the prevention of thrombus formation and/or its extension with a minimum risk of bleeding. The inhibition of a variety of proteolytic processes, particularly those of the coagulation cascade, has been reported as a property of plant protease inhibitors. The role of trypsin inhibitors (TIs) from Delonix regia (Dr) and Acacia schweinfurthii (As), members of the Kunitz family of protease inhibitors, was investigated on blood coagulation, platelet aggregation, and thrombus formation. Different from Acacia schweinfurthii trypsin inhibitor (AsTI), Delonix regia trypsin inhibitor (DrTI) is a potent inhibitor of FXIa with a Kiapp of 1.3 × 10-9 M. In vitro, both inhibitors at 100 µg corresponding to the concentrations of 21 µM and 15.4 µM of DrTI and AsTI, respectively, increased approximately 2.0 times the activated partial thromboplastin time (aPTT) in human plasma compared to the control, likely due to the inhibition of human plasma kallikrein (huPK) or activated factor XI (FXIa), in the case of DrTI. Investigating in vivo models of arterial thrombus formation and bleeding time, DrTI and AsTI, 1.3 µM and 0.96 µM, respectively, prolonged approximately 50% the time for total carotid artery occlusion in mice compared to the control. In contrast to heparin, the bleeding time in mice treated with the two inhibitors did not differ from that of the control group. DrTI and AsTI inhibited 49.3% and 63.8%, respectively, ex vivo murine platelet aggregation induced by adenosine diphosphate (ADP), indicating that these protein inhibitors prevent arterial thrombus formation possibly by interfering with the plasma kallikrein (PK) proteolytic action on the intrinsic coagulation pathway and its ability to enhance the platelet aggregation activity on the intravascular compartment leading to the improvement of a thrombus.


Assuntos
Plantas/química , Calicreína Plasmática/metabolismo , Inibidores de Proteases/uso terapêutico , Trombose/tratamento farmacológico , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Inibidores de Proteases/farmacologia
7.
EBioMedicine ; 37: 535-546, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30348621

RESUMO

BACKGROUND: The mechanism behind an increased risk of recurrent pregnancy loss (RPL) remains largely unknown. In our previous study, we identified that inter-α-trypsin inhibitor heavy chain 4 (ITI-H4) is highly expressed at a modified molecular weight of 36 kDa in serum derived from RPL patients. Yet, the precise molecular mechanism and pathways by which the short form of ITI-H4 carries out its function remain obscure. METHODS: Human sera and peripheral blood mononucleated cells (PBMCs) were collected from patients and normal controls to compare the expression levels of ITI-H4 and plasma kallikrein (KLKB1). Flow cytometric assay was performed to measure inflammatory markers in sera and culture supernatants. Furthermore, to investigate the functions of the two isoforms of ITI-H4, we performed migration, invasion, and proliferation assays. FINDINGS: In the current study, we showed that ITI-H4 as a biomarker of RPL could be regulated by KLKB1 through the IL-6 signaling cascade, indicating a novel regulatory system for inflammation in RPL. In addition, our study indicates that the two isoforms of ITI-H4 possess opposing functions on immune response, trophoblast invasion, and monocytes migration or proliferation. INTERPRETATION: The ITI-H4 (∆N688) might be a crucial inflammatory factor which contributes to the pathogenesis of RPL. Moreover, it is expected that this study would give some insights into potential functional mechanisms underlying RPL. FUND: This study was supported by the Ministry of Health & Welfare of the Republic of Korea (HI18C0378) through the Korea Health Industry Development Institute.


Assuntos
Aborto Habitual/sangue , Movimento Celular , Proliferação de Células , Glicoproteínas/sangue , Monócitos/metabolismo , Proteínas Secretadas Inibidoras de Proteinases/sangue , Transdução de Sinais , Trofoblastos/metabolismo , Aborto Habitual/patologia , Adulto , Biomarcadores/sangue , Proteínas Sanguíneas , Feminino , Humanos , Interleucina-6/sangue , Monócitos/patologia , Calicreína Plasmática/metabolismo , Gravidez , Fatores de Risco , Trofoblastos/patologia
8.
Biochem Biophys Res Commun ; 504(4): 857-864, 2018 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-30219233

RESUMO

Acute liver injury (ALI) is highly lethal acute liver failure caused by different etiologies. Transforming growth factor ß (TGF-ß) is a multifunctional cytokine and a well-recognized inducer of apoptotic and necrotic cell death in hepatocytes. Latent TGF-ß is activated partly through proteolytic cleavage by a serine protease plasma kallikrein (PLK) between the R58 and L59 residues of its propeptide region. Recently, we developed a specific monoclonal antibody to detect the N-terminal side LAP degradation products ending at residue R58 (R58 LAP-DPs) that reflect PLK-dependent TGF-ß activation. This study aimed to explore the potential roles of PLK-dependent TGF-ß activation in the pathogenesis of ALI. We established a mouse ALI model via the injection of anti-Fas antibodies (Jo2) and observed increases in the TGF-ß1 mRNA level, Smad3 phosphorylation, TUNEL-positive apoptotic hepatocytes and R58-positive cells in the liver tissues of Jo2-treated mice. The R58 LAP-DPs were observed in/around F4/80-positive macrophages, while macrophage depletion with clodronate liposomes partly alleviated the Jo2-induced liver injury. Blocking PLK-dependent TGF-ß activation using either the serine proteinase inhibitor FOY305 or the selective PLK inhibitor PKSI-527 or blocking the TGF-ß receptor-mediated signaling pathway using SB431542 significantly prevented Jo2-induced hepatic apoptosis and mortality. Furthermore, similar phenomena were observed in the mouse model of ALI with the administration of acetaminophen (APAP). In summary, R58 LAP-DPs reflecting PLK-dependent TGF-ß activation may serve as a biomarker for ALI, and targeting PLK-dependent TGF-ß activation has potential as a therapeutic strategy for ALI.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/prevenção & controle , Calicreína Plasmática/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Acetaminofen/efeitos adversos , Lesão Pulmonar Aguda/tratamento farmacológico , Animais , Anticorpos Monoclonais/efeitos adversos , Benzamidas/farmacologia , Biomarcadores/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Dioxóis/farmacologia , Modelos Animais de Doenças , Proteínas de Ligação a TGF-beta Latente/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Calicreína Plasmática/genética , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/genética , Receptor fas/imunologia
9.
Cell Signal ; 51: 257-265, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30118759

RESUMO

Factor XII (FXII) is a protease that is mainly produced in the liver and circulates in plasma as a single chain zymogen. Following contact with negatively charged surfaces, FXII is converted into the two-chain active form, FXIIa. FXIIa initiates the intrinsic blood coagulation pathway via activation of factor XI. Furthermore, it converts plasma prekallikrein to kallikrein (PK), which reciprocally activates FXII and liberates bradykinin from high molecular weight kininogen. In addition, FXIIa initiates fibrinolysis via PK-mediated urokinase activation and activates the classical complement pathway. Even though the main function of FXII seems to relate to the activation of the intrinsic coagulation pathway and the kallikrein-kinin system, a growing body of evidence suggests that FXII may also directly regulate cellular responses. In this regard, it has been found that FXII/FXIIa induces the expression of inflammatory mediators, promotes cell proliferation, and enhances the migration of neutrophils and lung fibroblasts. In addition, it has been reported that genetic ablation of FXII protects against neuroinflammation, reduces the formation of atherosclerotic lesions in Apoe-/- mice, improves wound healing, and inhibits postnatal angiogenesis. Although the aforementioned effects can be partially explained by the downstream products of FXII activation, the ability of FXII/FXIIa to directly regulate cellular responses has recently emerged as an alternative hypothesis. These direct cellular reactions to FXII/FXIIa will be discussed in the review.


Assuntos
Coagulação Sanguínea/imunologia , Fator XII/química , Fator XII/fisiologia , Inflamação , Animais , Aterosclerose/imunologia , Bradicinina/metabolismo , Movimento Celular , Proliferação de Células , Via Clássica do Complemento/imunologia , Fator XI/metabolismo , Fibrinólise/imunologia , Fibroblastos/imunologia , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Cininogênio de Alto Peso Molecular/metabolismo , Camundongos , Neutrófilos/imunologia , Calicreína Plasmática/metabolismo , Pré-Calicreína/metabolismo , Cicatrização/imunologia
10.
FASEB J ; 31(12): 5419-5431, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28808141

RESUMO

The plasma kallikrein-kinin system (KKS) consists of serine proteases, prekallikrein (pKal) and factor XII (FXII), and a cofactor, high-MW kininogen (HK). Upon activation, activated pKal and FXII cleave HK to release bradykinin. Activation of this system has been noted in patients with rheumatoid arthritis, and its pathogenic role has been characterized in animal arthritic models. In this study, we generated 2 knockout mouse strains that lacked pKal and HK and determined the role of KKS in autoantibody-induced arthritis. In a K/BxN serum transfer-induced arthritis (STIA) model, mice that lacked HK, pKal, or bradykinin receptors displayed protective phenotypes in joint swelling, histologic changes in inflammation, and cytokine production; however, FXII-deficient mice developed normal arthritis. Inhibition of Kal ameliorated arthritis severity and incidence at early stage STIA and reduced the levels of major cytokines in joints. In addition to releasing bradykinin from HK, Kal directly activated monocytes to produce proinflammatory cytokines, up-regulated their C5aR and FcRIII expression, and released C5a. Immune complex increased pKal activity, which led to HK cleavage. The absence of HK is associated with a decrease in joint vasopermeability. Thus, we identify a critical role for Kal in autoantibody-induced arthritis with pleiotropic effects, which suggests that it is a new target for the inhibition of arthritis.-Yang, A., Zhou, J., Wang, B., Dai, J., Colman, R. W., Song, W., Wu, Y. A critical role for plasma kallikrein in the pathogenesis of autoantibody-induced arthritis.


Assuntos
Artrite/metabolismo , Artrite/patologia , Autoanticorpos/metabolismo , Calicreína Plasmática/metabolismo , Animais , Artrite/genética , Artrite/imunologia , Bradicinina/metabolismo , Citocinas/metabolismo , Fator XII/genética , Fator XII/metabolismo , Feminino , Humanos , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Masculino , Camundongos , Camundongos Knockout , Monócitos/metabolismo , Calicreína Plasmática/genética , Reação em Cadeia da Polimerase
11.
Curr Opin Hematol ; 24(5): 411-418, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28604413

RESUMO

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


Assuntos
Fator XIIa/metabolismo , Calicreína Plasmática/metabolismo , Pré-Calicreína/metabolismo , Fator XIIa/genética , Humanos , Calicreína Plasmática/genética , Pré-Calicreína/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
13.
Int J Gynecol Cancer ; 26(8): 1366-74, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27465901

RESUMO

OBJECTIVE: At present, considerable efforts have been made to identify new cancer-specific markers for ovarian cancer (OC) diagnosis and the kallikrein-related peptidases (KLKs) family is one of the most studied candidates. This meta-analysis aims to evaluate the pooled diagnostic value of serum KLK measurement for diagnosing OC. METHODS: The Cochrane Library, PubMed, Excerpt Medica Database were searched for all relevant literature. The Quality Assessment for Studies of Diagnostic Accuracy tool was applied to assess the quality of enrolled studies. Statistical analysis was conducted by using Stata 13.0 software and Meta-Disc. RESULTS: A total of 15 studies from 13 articles were considered eligible for inclusion in the present analysis. The following pooled parameters were calculated by using the bivariate model: sensitivity of 0.582 (95% confidence interval [CI], 0.517-0.644), specificity of 0.909 (95% CI, 0.833-0.952), positive likelihood ratios of 6.367 (95% CI, 3.330-12.172), negative likelihood ratios of 0.460 (95% CI, 0.388-0.546), diagnostic odds ratio of 13.831 (95% CI, 6.460-29.614), respectively. CONCLUSIONS: Kallikrein-related peptidase seems to be a promising candidate biomarker in diagnosing OC, but the associated poor sensitivity of KLK individually may limit its value in clinical application. To resolve this problem, the combination of KLK and other markers may offer improved performance than a single marker.


Assuntos
Biomarcadores Tumorais/sangue , Neoplasias Ovarianas/enzimologia , Calicreína Plasmática/metabolismo , Feminino , Humanos , Neoplasias Ovarianas/sangue , Neoplasias Ovarianas/diagnóstico , Ensaios Clínicos Controlados Aleatórios como Assunto
14.
Invest Ophthalmol Vis Sci ; 57(6): 2390-9, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-27138737

RESUMO

PURPOSE: Plasma kallikrein is a serine protease and circulating component of inflammation, which exerts clinically significant effects on vasogenic edema. This study examines the role of plasma kallikrein in VEGF-induced retinal edema. METHODS: Intravitreal injections of VEGF and saline vehicle were performed in plasma prekallikrein-deficient (KLKB1-/-) and wild-type (WT) mice, and in both rats and mice receiving a selective plasma kallikrein inhibitor, VA999272. Retinal vascular permeability (RVP) and retinal thickness were measured by Evans blue permeation and optical coherence tomography, respectively. The retinal kallikrein kinin system was examined by Western blotting and immunohistochemistry. Retinal neovascularization was investigated in KLKB1-/- and WT mice subjected to oxygen-induced retinopathy. RESULTS: Vascular endothelial growth factor-induced RVP and retinal thickening were reduced in KLKB1-/- mice by 68% and 47%, respectively, compared to VEGF responses in WT mice. Plasma kallikrein also contributes to TNFα-induced retinal thickening, which was reduced by 52% in KLKB1-/- mice. Systemic administration of VA999272 reduced VEGF-induced retinal thickening by 57% (P < 0.001) in mice and 53% (P < 0.001) in rats, compared to vehicle-treated controls. Intravitreal injection of VEGF in WT mice increased plasma prekallikrein in the retina, which was diffusely distributed throughout the inner and outer retinal layers. Avascular and neovascular areas induced by oxygen-induced retinopathy were similar in WT and KLKB1-/- mice. CONCLUSIONS: Vascular endothelial growth factor increases extravasation of plasma kallikrein into the retina, and plasma kallikrein is required for the full effects of VEGF on RVP and retinal thickening in rodents. Systemic plasma kallikrein inhibition may provide a therapeutic opportunity to treat VEGF-induced retina edema.


Assuntos
Edema Macular/metabolismo , Calicreína Plasmática/metabolismo , Retina/patologia , Animais , Western Blotting , Permeabilidade Capilar , Injeções Intravítreas , Edema Macular/induzido quimicamente , Edema Macular/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Calicreína Plasmática/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Retina/metabolismo , Retina/fisiopatologia , Tomografia de Coerência Óptica , Fator A de Crescimento do Endotélio Vascular/administração & dosagem , Fator A de Crescimento do Endotélio Vascular/intoxicação
15.
Pulm Med ; 2016: 9425807, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28044105

RESUMO

Inflammation is an essential process in many pulmonary diseases in which kinins are generated by protease action on kininogen, a phenomenon that is blocked by protease inhibitors. We evaluated kinin release in an in vivo lung inflammation model in rats, in the presence or absence of CeKI (C. echinata kallikrein inhibitor), a plasma kallikrein, cathepsin G, and proteinase-3 inhibitor, and rCeEI (recombinant C. echinata elastase inhibitor), which inhibits these proteases and also neutrophil elastase. Wistar rats were intravenously treated with buffer (negative control) or inhibitors and, subsequently, lipopolysaccharide was injected into their lungs. Blood, bronchoalveolar lavage fluid (BALF), and lung tissue were collected. In plasma, kinin release was higher in the LPS-treated animals in comparison to CeKI or rCeEI groups. rCeEI-treated animals presented less kinin than CeKI-treated group. Our data suggest that kinins play a pivotal role in lung inflammation and may be generated by different enzymes; however, neutrophil elastase seems to be the most important in the lung tissue context. These results open perspectives for a better understanding of biological process where neutrophil enzymes participate and indicate these plant inhibitors and their recombinant correlates for therapeutic trials involving pulmonary diseases.


Assuntos
Caesalpinia , Neutrófilos , Pneumonia , Animais , Catepsina G/metabolismo , Modelos Animais de Doenças , Cininogênios/metabolismo , Modelos Biológicos , Neutrófilos/efeitos dos fármacos , Neutrófilos/enzimologia , Compostos Fitoquímicos/farmacologia , Calicreína Plasmática/metabolismo , Pneumonia/tratamento farmacológico , Pneumonia/enzimologia , Inibidores de Proteases/farmacologia , Ratos , Sementes
16.
Breast Cancer Res ; 16(5): 433, 2014 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-25212966

RESUMO

INTRODUCTION: Interleukin-like epithelial-to-mesenchymal transition inducer (ILEI) is an essential cytokine in tumor progression that is upregulated in several cancers, and its altered subcellular localization is a predictor of poor survival in human breast cancer. However, the regulation of ILEI activity and the molecular meaning of its altered localization remain elusive. METHODS: The influence of serum withdrawal, broad-specificity protease inhibitors, different serine proteases and plasminogen depletion on the size and amount of the secreted ILEI protein was investigated by Western blot analysis of EpRas cells. Proteases with ILEI-processing capacity were identified by carrying out an in vitro cleavage assay. Murine mammary tumor and metastasis models of EpC40 and 4T1 cells overexpressing different mutant forms of ILEI were used-extended with in vivo aprotinin treatment for the inhibition of ILEI-processing proteases-to test the in vivo relevance of proteolytic cleavage. Stable knockdown of urokinase plasminogen activator receptor (uPAR) in EpRas cells was performed to investigate the involvement of uPAR in ILEI secretion. The subcellular localization of the ILEI protein in tumor cell lines was analyzed by immunofluorescence. Immunohistochemistry for ILEI localization and uPAR expression was performed on two human breast cancer arrays, and ILEI and uPAR scores were correlated with the metastasis-free survival of patients. RESULTS: We demonstrate that secreted ILEI requires site-specific proteolytic maturation into its short form for its tumor-promoting function, which is executed by serine proteases, most efficiently by plasmin. Noncleaved ILEI is tethered to fibronectin-containing fibers of the extracellular matrix through a propeptide-dependent interaction. In addition to ILEI processing, plasmin rapidly increases ILEI secretion by mobilizing its intracellular protein pool in a uPAR-dependent manner. Elevated ILEI secretion correlates with an altered subcellular localization of the protein, most likely representing a shift into secretory vesicles. Moreover, altered subcellular ILEI localization strongly correlates with high tumor cell-associated uPAR protein expression, as well as with poor survival, in human breast cancer. CONCLUSIONS: Our findings point out extracellular serine proteases, in particular plasmin, and uPAR as valuable therapeutic targets against ILEI-driven tumor progression and emphasize the prognostic relevance of ILEI localization and a combined ILEI-uPAR marker analysis in human breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Citocinas/fisiologia , Neoplasias Pulmonares/metabolismo , Proteínas de Neoplasias/fisiologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Progressão da Doença , Transição Epitelial-Mesenquimal , Feminino , Fibrinolisina/metabolismo , Humanos , Estimativa de Kaplan-Meier , Elastase de Leucócito/metabolismo , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/secundário , Camundongos Nus , Transplante de Neoplasias , Calicreína Plasmática/metabolismo , Processamento de Proteína Pós-Traducional , Transporte Proteico , Proteólise
17.
FASEB J ; 28(2): 923-34, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24249636

RESUMO

The soluble cleaved urokinase plasminogen activator receptor (scuPAR) is a circulating protein detected in multiple diseases, including various cancers, cardiovascular disease, and kidney disease, where elevated levels of scuPAR have been associated with worsening prognosis and increased disease aggressiveness. We aimed to identify novel genetic and biomolecular mechanisms regulating scuPAR levels. Elevated serum scuPAR levels were identified in asthma (n=514) and chronic obstructive pulmonary disease (COPD; n=219) cohorts when compared to controls (n=96). In these cohorts, a genome-wide association study of serum scuPAR levels identified a human plasma kallikrein gene (KLKB1) promoter polymorphism (rs4253238) associated with serum scuPAR levels in a control/asthma population (P=1.17 × 10(-7)), which was also observed in a COPD population (combined P=5.04 × 10(-12)). Using a fluorescent assay, we demonstrated that serum KLKB1 enzymatic activity was driven by rs4253238 and is inverse to scuPAR levels. Biochemical analysis identified that KLKB1 cleaves scuPAR and negates scuPAR's effects on primary human bronchial epithelial cells (HBECs) in vitro. Chymotrypsin was used as a proproteolytic control, while basal HBECs were used as a control to define scuPAR-driven effects. In summary, we reveal a novel post-translational regulatory mechanism for scuPAR using a hypothesis-free approach with implications for multiple human diseases.


Assuntos
Estudo de Associação Genômica Ampla/métodos , Calicreína Plasmática/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/sangue , Asma/sangue , Sítios de Ligação/genética , Western Blotting , Células Cultivadas , Haplótipos , Humanos , Desequilíbrio de Ligação/genética , Calicreína Plasmática/genética , Reação em Cadeia da Polimerase , Polimorfismo de Nucleotídeo Único/genética , Doença Pulmonar Obstrutiva Crônica/sangue , RNA Mensageiro/genética , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética
18.
Thromb Haemost ; 110(3): 399-407, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23846131

RESUMO

Plasma prekallikrein is the liver-derived precursor of the trypsin-like serine protease plasma kallikrein (PK) and circulates in plasma bound to high molecular weight kininogen. The zymogen is converted to PK by activated factor XII. PK drives multiple proteolytic reaction cascades in the cardiovascular system such as the intrinsic pathway of coagulation, the kallikrein-kinin system, the fibrinolytic system, the renin-angiotensin system and the alternative complement pathway. Here, we review the biochemistry and cell biology of PK and focus on recent in vivo studies that have established important functions of the protease in procoagulant and proinflammatory disease states. Targeting PK offers novel strategies not previously appreciated to interfere with thrombosis and vascular inflammation in a broad variety of diseases.


Assuntos
Bradicinina/metabolismo , Calicreína Plasmática/metabolismo , Animais , Aprotinina/química , Coagulação Sanguínea , Hemorragia Cerebral/metabolismo , Proteínas do Sistema Complemento , Cisteína/química , Retinopatia Diabética/metabolismo , Dissulfetos/química , Fator XIIa/química , Fibrinólise , Hemostasia , Humanos , Inflamação , Sistema Calicreína-Cinina , Calicreínas/química , Cininas/química , Camundongos , Oligonucleotídeos Antissenso/química , Peptídeos/química , Estrutura Terciária de Proteína , Proteólise , Sistema Renina-Angiotensina , Transdução de Sinais , Trombose/metabolismo , Tripsina/química
19.
Invest Ophthalmol Vis Sci ; 54(2): 1086-94, 2013 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-23299478

RESUMO

PURPOSE: Retinal hemorrhages occur in a variety of sight-threatening conditions including ocular trauma, high altitude retinopathy, and chronic diseases such as diabetic and hypertensive retinopathies. The goal of this study is to investigate the effects of blood in the vitreous on retinal vascular function in rats. METHODS: Intravitreal injections of autologous blood, plasma kallikrein (PK), bradykinin, and collagenase were performed in Sprague-Dawley and Long-Evans rats. Retinal vascular permeability was measured using vitreous fluorophotometry and Evans blue dye permeation. Leukostasis was measured by fluorescein isothiocyanate-coupled concanavalin A lectin and acridine orange labeling. Retinal hemorrhage was examined on retinal flatmounts. Primary cultures of bovine retinal pericytes were cultured in the presence of 25 nM PK for 24 hours. The pericyte-conditioned medium was collected and the collagen proteome was analyzed by tandem mass spectrometry. RESULTS: Intravitreal injection of autologous blood induced retinal vascular permeability and retinal leukostasis, and these responses were ameliorated by PK inhibition. Intravitreal injections of exogenous PK induced retinal vascular permeability, leukostasis, and retinal hemorrhage. Proteomic analyses showed that PK increased collagen degradation in pericyte-conditioned medium and purified type IV collagen. Intravitreal injection of collagenase mimicked PK's effect on retinal hemorrhage. CONCLUSIONS: Intraocular hemorrhage increases retinal vascular permeability and leukostasis, and these responses are mediated, in part, via PK. Intravitreal injections of either PK or collagenase, but not bradykinin, induce retinal hemorrhage in rats. PK exerts collagenase-like activity that may contribute to blood-retinal barrier dysfunction.


Assuntos
Calicreína Plasmática/metabolismo , Doenças Retinianas/etiologia , Hemorragia Retiniana/complicações , Vasos Retinianos/patologia , Animais , Sangue , Barreira Hematorretiniana/efeitos dos fármacos , Bradicinina/farmacologia , Permeabilidade Capilar , Bovinos , Células Cultivadas , Colagenases/farmacologia , Concanavalina A/metabolismo , Azul Evans/metabolismo , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/metabolismo , Fluorofotometria , Injeções Intravítreas , Leucostasia/etiologia , Masculino , Pericitos/efeitos dos fármacos , Pericitos/metabolismo , Calicreína Plasmática/farmacologia , Ratos , Ratos Long-Evans , Ratos Sprague-Dawley , Doenças Retinianas/metabolismo , Hemorragia Retiniana/metabolismo , Vasos Retinianos/metabolismo , Espectrometria de Massas em Tandem , Corpo Vítreo/efeitos dos fármacos , Corpo Vítreo/metabolismo
20.
Vestn Oftalmol ; 128(4): 78-81, 2012.
Artigo em Russo | MEDLINE | ID: mdl-22994115

RESUMO

Multifactor etiology of diabetic retinopathy (DR) determines difficulty of understanding of pathogenesis and need of search of effective approaches to study key mechanisms of development of this microvascular complication of diabetes mellitus (DM). Significant achievements of the last years show the contribution of two proteolytic systems into pathogenesis of DR, that control vascular tone and permeability - kallikrein-kinin (KKS) and renin-angiotensin systems (RAS). Among new approaches to DR treatment one of the most appropriate is an influence on KKS by means of inhibiting kallikrein, that leads to reduction of retinal vascular permeability and allows to prevent the development of macula oedema and other consequences of vascular wall damage in DR.


Assuntos
Permeabilidade Capilar/efeitos dos fármacos , Retinopatia Diabética , Sistema Calicreína-Cinina , Edema Macular/prevenção & controle , Terapia de Alvo Molecular/tendências , Calicreína Plasmática , Retinopatia Diabética/complicações , Retinopatia Diabética/tratamento farmacológico , Retinopatia Diabética/metabolismo , Descoberta de Drogas , Previsões , Humanos , Sistema Calicreína-Cinina/efeitos dos fármacos , Sistema Calicreína-Cinina/fisiologia , Edema Macular/etiologia , Edema Macular/metabolismo , Calicreína Plasmática/antagonistas & inibidores , Calicreína Plasmática/metabolismo , Sistema Renina-Angiotensina/fisiologia , Vasopressinas/antagonistas & inibidores , Vasopressinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA