Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros











Intervalo de ano de publicação
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.
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.
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
5.
J Med Chem ; 61(7): 2823-2836, 2018 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-29517911

RESUMO

Plasma kallikrein, a member of the kallikrein-kinin system, catalyzes the release of the bioactive peptide bradykinin, which induces inflammation, vasodilation, vessel permeability, and pain. Preclinical evidence implicates the activity of plasma kallikrein in diabetic retinopathy, which is a leading cause of visual loss in patients suffering from diabetes mellitus. Employing a technology based on phage-display combined with chemical cyclization, we have identified highly selective bicyclic peptide inhibitors with nano- and picomolar potencies toward plasma kallikrein. Stability in biological matrices was either intrinsic to the peptide or engineered via the introduction of non-natural amino acids and nonpeptidic bonds. The peptides prevented bradykinin release in vitro, and in vivo efficacy was demonstrated in both a rat paw edema model and in rodent models of diabetes-induced retinal permeability. With a highly extended half-life of ∼40 h in rabbit eyes following intravitreal administration, the bicyclic peptides are promising novel agents for the treatment of diabetic retinopathy and diabetic macular edema.


Assuntos
Compostos Bicíclicos com Pontes/síntese química , Compostos Bicíclicos com Pontes/farmacologia , Complicações do Diabetes/tratamento farmacológico , Retinopatia Diabética/tratamento farmacológico , Edema Macular/tratamento farmacológico , Edema Macular/etiologia , Calicreína Plasmática/antagonistas & inibidores , Inibidores de Proteases/síntese química , Inibidores de Proteases/farmacologia , Animais , Bradicinina/metabolismo , Edema/tratamento farmacológico , Olho/metabolismo , Pé/patologia , Meia-Vida , Injeções Intravítreas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Permeabilidade , Inibidores de Proteases/administração & dosagem , Coelhos , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade , Especificidade por Substrato , Corpo Vítreo/química , Corpo Vítreo/metabolismo
6.
Nat Commun ; 8: 16092, 2017 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-28714475

RESUMO

The rapid renal clearance of peptides in vivo limits this attractive platform for the treatment of a broad range of diseases that require prolonged drug half-lives. An intriguing approach for extending peptide circulation times works through a 'piggy-back' strategy in which peptides bind via a ligand to the long-lived serum protein albumin. In accordance with this strategy, we developed an easily synthesized albumin-binding ligand based on a peptide-fatty acid chimera that has a high affinity for human albumin (Kd=39 nM). This ligand prolongs the elimination half-life of cyclic peptides in rats 25-fold to over seven hours. Conjugation to a peptide factor XII inhibitor developed for anti-thrombotic therapy extends the half-life from 13 minutes to over five hours, inhibiting coagulation for eight hours in rabbits. This high-affinity albumin ligand could potentially extend the half-life of peptides in human to several days, substantially broadening the application range of peptides as therapeutics.


Assuntos
Portadores de Fármacos/química , Ácidos Graxos/química , Peptídeos/administração & dosagem , Albumina Sérica Humana/química , Acilação , Animais , Portadores de Fármacos/metabolismo , Portadores de Fármacos/farmacocinética , Fator XII/antagonistas & inibidores , Ácidos Graxos/metabolismo , Ácidos Graxos/farmacocinética , Meia-Vida , Humanos , Ligantes , Taxa de Depuração Metabólica , Peptídeos/metabolismo , Peptídeos/farmacocinética , Peptídeos Cíclicos/administração & dosagem , Peptídeos Cíclicos/metabolismo , Peptídeos Cíclicos/farmacocinética , Calicreína Plasmática/antagonistas & inibidores , Ligação Proteica , Coelhos , Ratos , Albumina Sérica Humana/metabolismo , Albumina Sérica Humana/farmacocinética , Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores
7.
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
8.
J Med Chem ; 58(22): 8868-76, 2015 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-26536069

RESUMO

All serine proteases hydrolyze peptide bonds by the same basic mechanism and have very similar active sites, in spite of the fact that individual proteases have different physiological functions. We here report a strategy for designing high-affinity and high-specificity serine protease inhibitors using a versatile peptide scaffold, a 10-mer peptide, mupain-1 (CPAYSRYLDC). Mupain-1 was previously reported as a specific inhibitor of murine urokinase-type plasminogen activator (Ki = 0.55 µM) without measurable affinity to plasma kallikrein (Ki > 1000 µM). On the basis of a structure-based rational design, we substituted five residues of mupain-1 and converted it to a potent plasma kallikrein inhibitor (Ki = 0.014 µM). X-ray crystal structure analysis showed that the new peptide was able to adapt a new set of enzyme surface interactions by a slightly changed backbone conformation. Thus, with an appropriate re-engineering, mupain-1 can be redesigned to specific inhibitors of other serine proteases.


Assuntos
Proteínas Sanguíneas/síntese química , Proteínas Sanguíneas/farmacologia , Peptídeos/química , Calicreína Plasmática/antagonistas & inibidores , Inibidores de Serina Proteinase/síntese química , Inibidores de Serina Proteinase/farmacologia , Desenho de Fármacos , Humanos , Cinética , Modelos Moleculares , Conformação Molecular , Mutagênese Sítio-Dirigida , Peptídeos Cíclicos/química , Peptídeos Cíclicos/farmacologia , Proteínas Recombinantes/química , Relação Estrutura-Atividade , Especificidade por Substrato
9.
Planta Med ; 79(3-4): 227-35, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23345168

RESUMO

In cancer tumors, growth, invasion, and formation of metastasis at a secondary site play a pivotal role, participating in diverse processes in the development of the pathology, such as degradation of extracellular matrix. Bauhinia seeds contain relatively large quantities of peptidase inhibitors, and two Bauhinia inhibitors were obtained in a recombinant form from the Bauhinia bauhinioides species, B. bauhinoides cruzipain inhibitor, which is a cysteine and serine peptidase inhibitor, and B. bauhinioides kallikrein inhibitor, which is a serine peptidase inhibitor. While recombinant B. bauhinoides cruzipain inhibitor inhibits human neutrophil elastase cathepsin G and the cysteine proteinase cathepsin L, recombinant B. bauhinioides kallikrein inhibitor inhibits plasma kallikrein and plasmin. The effects of recombinant B. bauhinoides cruzipain inhibitor and recombinant B. bauhinioides kallikrein inhibitor on the viability of tumor cell lines with a distinct potential of growth from the same tissue were compared to those of the clinical cytotoxic drug 5-fluorouracil. At 12.5 µM concentration, recombinant B. bauhinoides cruzipain inhibitor and recombinant B. bauhinioides kallikrein inhibitor were more efficient than 5-fluorouracil in inhibiting MKN-28 and Hs746T (gastric), HCT116 and HT29 (colorectal), SkBr-3 and MCF-7 (breast), and THP-1 and K562 (leukemia) cell lines. Additionally, recombinant B. bauhinoides cruzipain inhibitor inhibited 40 % of the migration of Hs746T, the most invasive gastric cell line, while recombinant B. bauhinioides kallikrein inhibitor did not affect cell migration. Recombinant B. bauhinioides kallikrein inhibitor and recombinant B. bauhinoides cruzipain inhibitor, even at high doses, did not affect hMSC proliferation while 5-fluorouracil greatly reduced the proliferation rates of hMSCs. Therefore, both recombinant B. bauhinoides cruzipain inhibitor and recombinant B. bauhinioides kallikrein inhibitor might be considered for further studies to block peptidase activities in order to target specific peptidase-mediated growth and invasion characteristics of individual tumors, mainly in patients resistant to 5-fluorouracil chemotherapy.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Bauhinia/química , Neoplasias/tratamento farmacológico , Inibidores de Proteases/farmacologia , Proteínas Recombinantes/farmacologia , Sementes/química , Catepsina G/antagonistas & inibidores , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Cisteína Endopeptidases/metabolismo , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Fluoruracila/farmacologia , Humanos , Neoplasias/patologia , Calicreína Plasmática/antagonistas & inibidores , Proteínas de Protozoários , Proteínas Recombinantes/genética
10.
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
11.
J Pept Sci ; 18(10): 620-5, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22961872

RESUMO

Plasmin is best known as the key molecule in the fibrinolytic system, which is critical for clot lysis and can initiate matrix metalloproteinase (MMP) activation cascade. Along with MMP, plasmin is suggested to be involved in physiological processes that are linked to the risk of carcinoma formation. Plasmin inhibitors could be perceived as a promising new principle in the treatment of diseases triggered by plasmin. On the basis of the peptidic sequence derived from the synthetic plasmin substrate, a series of peptidic plasmin inhibitors possessing nitrile as warhead were prepared and evaluated for their inhibitory activities against plasmin and other serine proteases, plasma kallikrein and urokinase. The most potent peptidic inhibitors with the nitrile warhead exhibit the potency toward plasmin (IC(50) = 7.7-11 µM) and are characterized by their selectivity profile against plasma kallikrein and urokinase. The results and molecular modeling of the peptidic inhibitor complexed with plasmin reveal that the P2 residue makes favorable contacts with the open binding pocket comprising the S2 and S3 subsites of plasmin.


Assuntos
Fibrinolisina/antagonistas & inibidores , Nitrilas/química , Oligopeptídeos/farmacologia , Inibidores de Serina Proteinase/química , Inibidores de Serina Proteinase/farmacologia , Relação Dose-Resposta a Droga , Humanos , Modelos Moleculares , Estrutura Molecular , Oligopeptídeos/síntese química , Oligopeptídeos/química , Calicreína Plasmática/antagonistas & inibidores , Inibidores de Serina Proteinase/síntese química , Relação Estrutura-Atividade , Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores
12.
Chembiochem ; 13(7): 1032-8, 2012 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-22492661

RESUMO

Combinatorial libraries of structurally diverse peptide macrocycles offer a rich source for the development of high-affinity ligands to targets of interest. In this work we have developed linkers for the generation of genetically encoded bicyclic peptides and tested whether the peptides cyclised by them have significant variations in their backbone conformations. Two new cyclisation reagents, each containing three thiol-reactive groups, efficiently and selectively cyclised linear peptides containing three cysteine moieties. When the mesitylene linker of the bicyclic peptide PK15, a potent inhibitor of plasma kallikrein (K(i)=2 nM), was replaced by the new linkers, its inhibitory activity dropped by a factor of more than 1000, suggesting that the linkers impose different conformations on the peptide. Indeed, structural analysis by solution-state NMR revealed different NOE constraints in the three bicyclic peptides, indicating that these relatively small linkers at the centres of bicyclic peptide structures significantly influence the conformations of the peptides. These results demonstrate the prominent structural role of linkers in peptide macrocycles and suggest that application of different cyclisation linkers in a combinatorial fashion could be an attractive means to generate topologically diverse macrocycle libraries.


Assuntos
Peptídeos Cíclicos/química , Peptídeos/química , Sequência de Aminoácidos , Técnicas de Química Combinatória , Ciclização , Humanos , Dados de Sequência Molecular , Estrutura Molecular , Biblioteca de Peptídeos , Peptídeos Cíclicos/síntese química , Calicreína Plasmática/antagonistas & inibidores , Calicreína Plasmática/química
13.
Drugs Today (Barc) ; 46(8): 547-55, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20830315

RESUMO

Hereditary angioedema (HAE) is a debilitating, potentially fatal disease characterized by variable and unpredictable acute attacks of swelling affecting the subcutaneous tissue and mucosa. It is an autosomal dominant disorder resulting from a genetic deficiency of functional C1-esterase inhibitor. Available treatments include long-term prophylaxis, short-term prophylaxis and treatment of acute attacks. Ecallantide is a novel, specific and potent inhibitor of plasma kallikrein that was recently approved in the United States for the treatment of acute attacks of HAE in patients aged 16 years and older. In two phase III clinical trials, the subcutaneous administration of 30 mg ecallantide resulted in significantly greater symptom improvement than placebo for acute attacks of HAE. Ecallantide was generally well tolerated throughout the clinical development program. The main safety concern following ecallantide treatment is hypersensitivity reactions, including anaphylaxis. A Risk Evaluation and Management Strategy (REMS) has been implemented to minimize this risk and a long-term observational safety study is currently under way to collect more information about hypersensitivity and immunogenicity. Ecallantide represents a novel treatment option for patients with HAE.


Assuntos
Angioedemas Hereditários/tratamento farmacológico , Inibidores Enzimáticos/uso terapêutico , Peptídeos/uso terapêutico , Calicreína Plasmática/antagonistas & inibidores , Angioedemas Hereditários/enzimologia , Hipersensibilidade a Drogas/etiologia , Inibidores Enzimáticos/efeitos adversos , Inibidores Enzimáticos/farmacocinética , Medicina Baseada em Evidências , Humanos , Peptídeos/efeitos adversos , Peptídeos/farmacocinética , Calicreína Plasmática/metabolismo , Medição de Risco , Resultado do Tratamento
14.
An Acad Bras Cienc ; 81(3): 615-21, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19722028

RESUMO

Obtained from leguminous seeds, various plant proteins inhibit animal proteinases, including human, and can be considered for the development of compounds with biological activity. Inhibitors from the Bowman-Birk and plant Kunitz-type family have been characterized by proteinase specificity, primary structure and reactive site. Our group mostly studies the genus Bauhinia, mainly the species bauhinioides, rufa, ungulata and variegata. In some species, more than one inhibitor was characterized, exhibiting different properties. Although proteins from this group share high structural similarity, they present differences in proteinase inhibition, explored in studies using diverse biological models.


Assuntos
Fabaceae/química , Inibidores de Proteases/farmacologia , Animais , Quimotripsina/antagonistas & inibidores , Fabaceae/classificação , Humanos , Peptídeos/isolamento & purificação , Peptídeos/farmacologia , Proteínas de Plantas/isolamento & purificação , Proteínas de Plantas/farmacologia , Calicreína Plasmática/antagonistas & inibidores , Inibidores de Proteases/isolamento & purificação , Sementes/química , Sementes/classificação , Inibidor da Tripsina de Soja de Bowman-Birk/isolamento & purificação , Inibidor da Tripsina de Soja de Bowman-Birk/farmacologia
15.
An. acad. bras. ciênc ; 81(3): 615-621, Sept. 2009. ilus, tab
Artigo em Inglês | LILACS | ID: lil-523985

RESUMO

Obtained from leguminous seeds, various plant proteins inhibit animal proteinases, including human, and can be considered for the development of compounds with biological activity. Inhibitors from the Bowman-Birk and plant Kunitz-type family have been characterized by proteinase specificity, primary structure and reactive site. Our group mostly studies the genus Bauhinia, mainly the species bauhinioides, rufa, ungulata and variegata. In some species, more than one inhibitor was characterized, exhibiting different properties. Although proteins from this group share high structural similarity, they present differences in proteinase inhibition, explored in studies using diverse biological models.


Obtidas de sementes leguminosas, várias proteínas inibem proteinases de origem animal, incluindo humanas, e podem ser consideradas para o desenvolvimento de compostos com atividade biológica. Inibidores da família Bowman-Birk e da família Kunitz vegetal tem sido caracterizados em relação a especificidade para proteinase, estrutura primária e sitio reativo. O nosso grupo majoritariamente vem estudando o gênero Bauhinia, principalmente as espécies bauhinioides, rufa, ungulatae variegata. Em algumas espécies, mais de um inibidor com propriedades diferentes foi caracterizado. Embora tais proteínas apresentem alta similaridade estrutural, diferem quanto à inibição de proteinases, e foram exploradas em estudos utilizando diversos modelos biológicos.


Assuntos
Animais , Humanos , Fabaceae/química , Inibidores de Proteases/farmacologia , Quimotripsina/antagonistas & inibidores , Fabaceae/classificação , Peptídeos/isolamento & purificação , Peptídeos/farmacologia , Proteínas de Plantas/isolamento & purificação , Proteínas de Plantas/farmacologia , Calicreína Plasmática/antagonistas & inibidores , Inibidores de Proteases/isolamento & purificação , Sementes/química , Sementes/classificação , Inibidor da Tripsina de Soja de Bowman-Birk/isolamento & purificação , Inibidor da Tripsina de Soja de Bowman-Birk/farmacologia
16.
Nat Chem Biol ; 5(7): 502-7, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19483697

RESUMO

Here we describe a phage strategy for the selection of ligands based on bicyclic or linear peptides attached covalently to an organic core. We designed peptide repertoires with three reactive cysteine residues, each spaced apart by several random amino acid residues, and we fused the repertoires to the phage gene-3-protein. Conjugation with tris-(bromomethyl)benzene via the reactive cysteines generated repertoires of peptide conjugates with two peptide loops anchored to a mesitylene core. Iterative affinity selections yielded several enzyme inhibitors; after further mutagenesis and selection, we were able to chemically synthesize a lead inhibitor (PK15; Ki =1.5 nM) specific to human plasma kallikrein that efficiently interrupted the intrinsic coagulation pathway in human plasma tested ex vivo. This approach offers a powerful means of generating and selecting bicyclic macrocycles (or if cleaved, linear derivatives thereof) as ligands poised at the interface of small-molecule drugs and biologics.


Assuntos
Técnicas de Química Combinatória , Inibidores Enzimáticos/química , Biblioteca de Peptídeos , Peptídeos Cíclicos/química , Sequência de Aminoácidos , Catepsina G/antagonistas & inibidores , Clonagem Molecular , Cisteína/química , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Humanos , Ligantes , Dados de Sequência Molecular , Peptídeos Cíclicos/síntese química , Peptídeos Cíclicos/genética , Peptídeos Cíclicos/farmacologia , Calicreína Plasmática/antagonistas & inibidores
17.
Biol Chem ; 387(8): 1129-38, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16895484

RESUMO

The interplay of different proteases and glycosaminoglycans is able to modulate the activity of the enzymes and to affect their structures. Human plasma kallikrein (huPK) is a proteolytic enzyme involved in intrinsic blood clotting, the kallikrein-kinin system and fibrinolysis. We investigated the effect of heparin on the action, inhibition and secondary structure of huPK. The catalytic efficiency for the hydrolysis of substrates by huPK was determined by Michaelis-Menten kinetic plots: 5.12x10(4) M-1 s-1 for acetyl-Phe-Arg-p-nitroanilide, 1.40x10(5) M-1 s-1 for H-D-Pro-Phe-Arg-p-nitroanilide, 2.25x10(4) M-1 s-1 for Abz-Gly-Phe-Ser-Pro-Phe-Arg-Ser-Ser-Arg-Gln-EDDnp, 4.24x10(2)M-1 s-1 for factor XII and 5.58x10(2) M-1 s-1 for plasminogen. Heparin reduced the hydrolysis of synthetic substrates (by 2.0-fold), but enhanced factor XII and plasminogen hydrolysis (7.7- and 1.4-fold, respectively). The second-order rate constants for inhibition of huPK by antithrombin and C1-inhibitor were 2.40x10(2) M-1 s-1 and 1.70x10(4) M-1 s-1, respectively. Heparin improved the inhibition of huPK by these inhibitors (3.4- and 1.4-fold). Despite the fact that huPK was able to bind to a heparin-Sepharose matrix, its secondary structure was not modified by heparin, as monitored by circular dichroism. These actions may have a function in the control or maintenance of some pathophysiological processes in which huPK participates.


Assuntos
Inibidores Enzimáticos/farmacologia , Heparina/farmacologia , Peptídeos/metabolismo , Calicreína Plasmática/antagonistas & inibidores , Calicreína Plasmática/metabolismo , Antitrombinas/farmacologia , Catálise , Proteína Inibidora do Complemento C1/farmacologia , Fator XII/efeitos dos fármacos , Fator XII/metabolismo , Humanos , Hidrólise , Peptídeos/efeitos dos fármacos , Calicreína Plasmática/química , Plasminogênio/efeitos dos fármacos , Plasminogênio/metabolismo , Estrutura Secundária de Proteína , Fatores de Tempo , alfa 1-Antitripsina/farmacologia
18.
J Mol Biol ; 346(5): 1335-49, 2005 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-15713485

RESUMO

Hepatocyte growth factor activator (HGFA) is a serine protease that converts hepatocyte growth factor (HGF) into its active form. When activated HGF binds its cognate receptor Met, cellular signals lead to cell growth, differentiation, and migration, activities which promote tissue regeneration in liver, kidney and skin. Intervention in the conversion of HGF to its active form has the potential to provide therapeutic benefit where HGF/Met activity is associated with tumorigenesis. To help identify ways to moderate HGF/Met effects, we have determined the molecular structure of the protease domain of HGFA. The structure we determined, at 2.7 A resolution, with no pseudo-substrate or inhibitor bound is characterized by an unconventional conformation of key residues in the enzyme active site. In order to find whether this apparently non-enzymatically competent arrangement would persist in the presence of a strongly-interacting inhibitor, we also have determined, at 2.6 A resolution, the X-ray structure of HGFA complexed with the first Kunitz domain (KD1) from the physiological inhibitor hepatocyte growth factor activator inhibitor 1B (HAI-1B). In this complex we observe a rearranged substrate binding cleft that closely mirrors the cleft of other serine proteases, suggesting an extreme conformational dynamism. We also characterize the inhibition of 16 serine proteases by KD1, finding that the previously reported enzyme specificity of the intact extracellular region of HAI-1B resides in KD1 alone. We find that HGFA, matriptase, hepsin, plasma kallikrein and trypsin are potently inhibited, and use the complex structure to rationalize the structural basis of these results.


Assuntos
Fator de Crescimento de Hepatócito/metabolismo , Glicoproteínas de Membrana/metabolismo , Conformação Proteica , Serina Endopeptidases , Sequência de Aminoácidos , Sítios de Ligação , Cristalografia por Raios X , Humanos , Dados de Sequência Molecular , Calicreína Plasmática/antagonistas & inibidores , Calicreína Plasmática/química , Calicreína Plasmática/metabolismo , Ligação Proteica , Proteínas Secretadas Inibidoras de Proteinases , Homologia de Sequência de Aminoácidos , Serina Endopeptidases/química , Serina Endopeptidases/metabolismo , Inibidores de Serina Proteinase/metabolismo , Especificidade por Substrato , Tripsina/química , Tripsina/metabolismo
19.
Artigo em Inglês | MEDLINE | ID: mdl-15638742

RESUMO

Bradykinin and Lys-bradykinin are potent peptide mediators implicated in several physiopathological effects in mammals. They act through activation of G-protein-coupled constitutive B(2) or inducible kinin B(1) receptors linked to signaling pathways involving increased intracellular Ca(++) concentrations and/or release of mediators including arachidonic acid metabolites, NO and EDHF. In the cardiovascular system, the kallikrein-kinin system exerts a fine control of vascular smooth muscle tone and arterial blood pressure, and plays a significant cardioprotective effect. This has been lately confirmed in experimental studies employing transgenic mice overexpressing human tissue kallikrein and animals with knockout of kinin B(1) and B(2) receptor gene. Disturbances in this system are associated with arterial hypertension, myocardial ischaemia and other clinical complications. Inhibitors of kininase II (angiotensin-converting enzyme) have been prescribed successfully to patients with cardiovascular diseases, but there is still a great interest in developing drugs or pharmacological strategies that augment the activity of kininogen-kallikrein-kinin system in pathological conditions. Delivery of adenovirus vector containing the human tissue kallikrein gene (gene kallikrein therapy) has emerged as a great potential to satisfy these conditions. This review provides a summary of plasma and tissue kallikrein-kinin system, focusing on the pharmacological properties, kinin receptors and drugs reported to interfere with their actions. The modulatory effects of the kallikrein-kinin system on cardiovascular system, particularly in regulating smooth muscle tone and arterial blood pressure and in preventing myocardium ischaemia have also been explored in the review.


Assuntos
Bradicinina/fisiologia , Sistema Cardiovascular/metabolismo , Sistema Calicreína-Cinina/fisiologia , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Animais , Bradicinina/biossíntese , Bradicinina/metabolismo , Cardiotônicos/uso terapêutico , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/metabolismo , Sistema Cardiovascular/efeitos dos fármacos , Terapia Genética , Humanos , Calidina/biossíntese , Calidina/metabolismo , Sistema Calicreína-Cinina/efeitos dos fármacos , Cininogênios/metabolismo , Cininas/antagonistas & inibidores , Cininas/genética , Cininas/metabolismo , Calicreína Plasmática/antagonistas & inibidores , Calicreína Plasmática/metabolismo , Inibidores de Proteases/uso terapêutico , Calicreínas Teciduais/antagonistas & inibidores , Calicreínas Teciduais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA