Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Ann Clin Lab Sci ; 51(6): 795-804, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34921033

RESUMO

OBJECTIVE: Lipoprotein (a) [Lp(a)] is an LDL-like particle constituted by lipids, apolipoprotein B100 and apolipoprotein (a) [apo(a)], a multidomain glycoprotein whose molecular mass is dependent on the genetically encoded number of Kringle IV type 2 (KIV-2) repeats. Because Lp(a) isoforms have been associated with cardiovascular risk (CVR), we have investigated if their interfacial properties can contribute to distinguish between low and high-risk groups and thus be used as a new CVR indicator. METHODS: Four Lp(a) variants, each carrying a different apo(a) isoform (K20, K24, K25, and K29), were purified from plasma of homozygous donors and their interfacial properties characterized using ellipsometry and surface pressure techniques. RESULTS: Ellipsometry measurements revealed that these isoforms had a similar propensity to form adsorbed layers at hydrophobic-hydrophilic interfaces, but surface pressure enabled to clearly separate them into two groups: K20 and K24 on one side, and K25 and K29 on the other side. CONCLUSION: Though K24 and K25 differ only by a single KIV-2 domain, their sharp difference in surface pressure suggests a critical threshold between the two Lp(a) forms, providing insights into the use of condensed matter approaches to monitor CVR. Our findings may represent a new laboratory window to assist medical decisions and to develop precision medicine treatments, practices, and products for CVR, which can be extended to other cardiovascular disease conditions.


Assuntos
Doenças Cardiovasculares , Lipoproteína(a) , Isoformas de Proteínas , Doenças Cardiovasculares/diagnóstico , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/terapia , Técnicas de Química Analítica/métodos , Fatores de Risco de Doenças Cardíacas , Humanos , Interações Hidrofóbicas e Hidrofílicas , Kringles/fisiologia , Metabolismo dos Lipídeos , Lipoproteína(a)/química , Lipoproteína(a)/metabolismo , Medicina de Precisão/métodos , Isoformas de Proteínas/química , Isoformas de Proteínas/classificação , Isoformas de Proteínas/isolamento & purificação , Propriedades de Superfície
2.
Biochem J ; 477(5): 953-970, 2020 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-32069359

RESUMO

To understand the role of substrate plasminogen kringles in its differential catalytic processing by the streptokinase - human plasmin (SK-HPN) activator enzyme, Fluorescence Resonance Energy Transfer (FRET) model was generated between the donor labeled activator enzyme and the acceptor labeled substrate plasminogen (for both kringle rich Lys plasminogen - LysPG, and kringle less microplasminogen - µPG as substrates). Different steps of plasminogen to plasmin catalysis i.e. substrate plasminogen docking to scissile peptide bond cleavage, chemical transformation into proteolytically active product, and the decoupling of the nascent product from the SK-HPN activator enzyme were segregated selectively using (1) FRET signal as a proximity sensor to score the interactions between the substrate and the activator during the cycle of catalysis, (2) active site titration studies and (3) kinetics of peptide bond cleavage in the substrate. Remarkably, active site titration studies and the kinetics of peptide bond cleavage have shown that post docking chemical transformation of the substrate into the product is independent of kringles adjacent to the catalytic domain (CD). Stopped-flow based rapid mixing experiments for kringle rich and kringle less substrate plasminogen derivatives under substrate saturating and single cycle turnover conditions have shown that the presence of kringle domains adjacent to the CD in the macromolecular substrate contributes by selectively speeding up the final step, namely the product release/expulsion step of catalysis by the streptokinase-plasmin(ogen) activator enzyme.


Assuntos
Domínio Catalítico/fisiologia , Fibrinolisina/metabolismo , Kringles/fisiologia , Plasminogênio/metabolismo , Estreptoquinase/metabolismo , Catálise , Fibrinolisina/química , Transferência Ressonante de Energia de Fluorescência/métodos , Humanos , Plasminogênio/química , Estrutura Secundária de Proteína , Estreptoquinase/química , Especificidade por Substrato/fisiologia
3.
J Chromatogr A ; 1401: 42-51, 2015 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-25981289

RESUMO

The interactions between angiogenesis inhibitor Kringle 5 and its five specific ligands were investigated by frontal affinity chromatography in combination with fluorescence spectra and site-directed molecular docking. The binding constants of trans-4-(aminomethyl) cyclohexane carboxylic acid (AMCHA), epsilon-aminocaproic acid (EACA), benzylamine, 7-aminoheptanoic acid (7-AHA) and L-lysine to Kringle 5 were 19.0×10(3), 7.97×10(3), 6.45×10(3), 6.07×10(3) and 4.04×10(3) L/mol, respectively. The five ligands bound to Kringle 5 on the lysine binding site in equimolar amounts, which was pushed mainly by hydrogen bond and Van der Waals force. This binding affinity was believed to be dependent on the functional group and flexible feature in ligands. This study will provide an important insight into the binding mechanism of angiogenesis inhibitor Kringle 5 to its specific ligands.


Assuntos
Cromatografia de Afinidade , Kringles/fisiologia , Inibidores da Angiogênese/metabolismo , Sítios de Ligação , Ligação de Hidrogênio , Ligantes , Lisina/química , Ligação Proteica
4.
Biochemistry ; 53(3): 505-17, 2014 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-24383758

RESUMO

Kunitz domain 1 (KD1) of tissue factor pathway inhibitor-2 in which P2' residue Leu17 (bovine pancreatic trypsin inhibitor numbering) is mutated to Arg selectively inhibits the active site of plasmin with ∼5-fold improved affinity. Thrombin cleavage (24 h extended incubation at a 1:50 enzyme-to-substrate ratio) of the KD1 mutant (Leu17Arg) yielded a smaller molecule containing the intact Kunitz domain with no detectable change in the active-site inhibitory function. The N-terminal sequencing and MALDI-TOF/ESI data revealed that the starting molecule has a C-terminal valine (KD1L17R-VT), whereas the smaller molecule has a C-terminal lysine (KD1L17R-KT). Because KD1L17R-KT has C-terminal lysine, we examined whether it could serve as a decoy receptor for plasminogen/plasmin. Such a molecule might inhibit plasminogen activation as well as the active site of generated plasmin. In surface plasmon resonance experiments, tissue plasminogen activator (tPA) and Glu-plasminogen bound to KD1L17R-KT (Kd ∼ 0.2 to 0.3 µM) but not to KD1L17R-VT. Furthermore, KD1L17R-KT inhibited tPA-induced plasma clot fibrinolysis more efficiently than KD1L17R-VT. Additionally, compared to ε-aminocaproic acid KD1L17R-KT was more effective in reducing blood loss in a mouse liver-laceration injury model, where the fibrinolytic system is activated. In further experiments, the micro(µ)-plasmin-KD1L17R-KT complex inhibited urokinase-induced plasminogen activation on phorbol-12-myristate-13-acetate-stimulated U937 monocyte-like cells, whereas the µ-plasmin-KD1L17R-VT complex failed to inhibit this process. In conclusion, KD1L17R-KT inhibits the active site of plasmin as well as acts as a decoy receptor for the kringle domain(s) of plasminogen/plasmin; hence, it limits both plasmin generation and activity. With its dual function, KD1L17R-KT could serve as a preferred agent for controlling plasminogen activation in pathological processes.


Assuntos
Fibrinolisina/metabolismo , Glicoproteínas/farmacologia , Kringles/fisiologia , Receptores de Superfície Celular/química , Sequência de Aminoácidos , Animais , Fibrinolisina/antagonistas & inibidores , Fibrinólise/efeitos dos fármacos , Glicoproteínas/genética , Humanos , Kringles/efeitos dos fármacos , Lacerações/tratamento farmacológico , Fígado/lesões , Camundongos , Plasminogênio/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Ativador de Plasminogênio Tecidual/antagonistas & inibidores , Células U937
5.
J Neurosci Res ; 88(7): 1537-48, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20025058

RESUMO

We have shown that prothrombin kringle-2 (pKr-2), a domain of human prothrombin distinct from thrombin could activate cultured rat brain microglia in vitro. However, little is known whether pKr-2-induced microglial activation could cause neurotoxicity on dopaminergic (DA) neurons in vivo. To address this question, pKr-2 was injected into the rat substantia nigra (SN). Tyrosine hydroxylase (TH) immunohistochemistry experiments demonstrate significant loss of DA neurons seven days after injection of pKr-2. In parallel, pKr-2-activated microglia were detected in the SN with OX-42 and OX-6 immunohistochemistry. Reverse transcription PCR and double-label immunohistochemistry revealed that activated microglia in vivo exhibit early and transient expression of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2) and several proinflammatory cytokines. The pKr-2-induced loss of SN DA neurons was partially inhibited by the NOS inhibitor N(G)-nitro-L-arginine methyl ester hydrochloride, and the COX-2 inhibitor DuP-697. Extracellular signal-regulated kinase 1/2, c-Jun N-terminal kinase and p38 mitogen-activated protein kinase were activated in the SN as early as 1 hr after pKr-2 injection, and localized within microglia. Inhibition of these kinases led to attenuation of mRNA expression of iNOS, COX-2 and several proinflammatory cytokines, and rescue of DA neurons in the SN. Intriguingly, following treatment with pKr-2 in vitro, neurotoxicity was detected exclusively in co-cultures of mesencephalic neurons and microglia, but not microglia-free neuron-enriched mesencephalic cultures, indicating that microglia are required for pKr-2 neurotoxicity. Our results strongly suggest that microglia activated by endogenous compound(s), such as pKr-2, are implicated in the DA neuronal cell death in the SN.


Assuntos
Dopamina/metabolismo , Gliose/metabolismo , Microglia/metabolismo , Neurônios/metabolismo , Protrombina/metabolismo , Substância Negra/metabolismo , Animais , Antígeno CD11b/análise , Antígeno CD11b/metabolismo , Células Cultivadas , Técnicas de Cocultura , Ciclo-Oxigenase 2/efeitos dos fármacos , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Inibidores de Ciclo-Oxigenase 2/farmacologia , Feminino , Gliose/induzido quimicamente , Gliose/fisiopatologia , Mediadores da Inflamação/metabolismo , Kringles/fisiologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Microglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Doença de Parkinson/metabolismo , Doença de Parkinson/fisiopatologia , Protrombina/química , Protrombina/toxicidade , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Substância Negra/efeitos dos fármacos , Substância Negra/fisiopatologia
6.
J Struct Biol ; 169(3): 349-59, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19800007

RESUMO

The solution structure of the complex containing the isolated kringle 2 domain of human plasminogen (K2(Pg)) and VEK-30, a 30-amino acid residue internal peptide from a streptococcal M-like plasminogen (Pg) binding protein (PAM), has been determined by multinuclear high-resolution NMR. Complete backbone and side-chain assignments were obtained from triple-resonance experiments, after which structure calculations were performed and ultimately refined by restrained molecular simulation in water. We find that, in contrast with the dimer of complexes observed in the asymmetric unit of the crystal, global correlation times and buoyant molecular weight determinations of the complex and its individual components showed the monomeric nature of all species in solution. The NMR-derived structure of K2(Pg) in complex with VEK-30 presents a folding pattern typical of other kringle domains, while bound VEK-30 forms an end-to-end alpha-helix (residues 6-27) in the complex. Most of the VEK-30/K2(Pg) interactions in solution occur between a single face of the alpha-helix of VEK-30 and the lysine binding site (LBS) of K2(Pg). The canonical LBS of K2(Pg), consisting of Asp54, Asp56, Trp60, Arg69, and Trp70 (kringle numbering), interacts with an internal pseudo-lysine of VEK-30, comprising side-chains of Arg17, His18, and Glu20. Site-specific mutagenesis analysis confirmed that the electrostatic field formed by the N-terminal anionic residues of the VEK-30 alpha-helix, viz., Asp7, and the non-conserved cationic residues of K2(Pg), viz., Lys43 and Arg55, play additional important roles in the docking of VEK-30 to K2(Pg). Structural analysis and kringle sequence alignments revealed several important features related to exosite binding that provide a structural rationale for the high specificity and affinity of VEK-30 for K2(Pg).


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Plasminogênio/química , Plasminogênio/metabolismo , Proteínas de Bactérias/genética , Proteínas de Transporte/genética , Dicroísmo Circular , Humanos , Kringles/genética , Kringles/fisiologia , Espectroscopia de Ressonância Magnética , Mutagênese Sítio-Dirigida , Plasminogênio/genética , Ligação Proteica/genética , Ligação Proteica/fisiologia , Ressonância de Plasmônio de Superfície , Ultracentrifugação
7.
Int J Obes (Lond) ; 33(10): 1136-42, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19636317

RESUMO

BACKGROUND: Lipoprotein(a) [Lp(a)] is considered an independent risk factor for cardiovascular disease. Its concentration is mainly determined by the kringle-IV repeat copy number variation (CNV) at the apolipoprotein(a) [apo(a)] locus. OBJECTIVE: We aimed to investigate the immediate effect of weight reduction on plasma Lp(a) levels and its dependency on the apo(a) CNV in obese children. DESIGN: We performed a prospective longitudinal intervention study of a low-fat hypocaloric diet conducted in a 3-week dietary camp for obese children. In all, 140 obese participants (54 boys and 86 girls) with a mean age of 12.5+/-1.6 years and a mean relative body mass index (BMI) before treatment of 165.6+/-24.7% were included. Body weight and plasma levels of Lp(a), lipids, apolipoproteins A-I and B, insulin, and C-reactive protein were determined before the onset and after the end of the intervention. In addition, the number of apo(a) kringle-IV repeats were determined using sodium dodecyl sulfate agarose gel electrophoresis. RESULTS: The mean loss of body weight was 5.0+/-1.3 kg (-6.6%), resulting in a mean decrease of the relative BMI of 6.6%. Blood chemistry revealed significant changes in all parameters, especially in Lp(a), with a decrease from 24.4+/-30.6 to 17.9+/-22.6 mg per 100 ml or -19% (P<0.001). The decrease of Lp(a) levels was higher in the group with low compared with high molecular weight apo(a) phenotypes (-23.9 vs -16.6%). CONCLUSIONS: Weight reduction in obese children is associated with significant changes in Lp(a) levels, especially in subjects with high pre-treatment Lp(a) concentrations. This effect is markedly influenced by the molecular phenotype at the copy-number variable apo(a) locus.


Assuntos
Apolipoproteínas A/sangue , Doenças Cardiovasculares/sangue , Kringles/fisiologia , Lipoproteína(a)/sangue , Obesidade/sangue , Redução de Peso/fisiologia , Adolescente , Apolipoproteínas A/genética , Índice de Massa Corporal , Doenças Cardiovasculares/genética , Criança , Dieta com Restrição de Gorduras , Feminino , Humanos , Kringles/genética , Lipoproteína(a)/genética , Estudos Longitudinais , Masculino , Obesidade/genética , Fenótipo , Estudos Prospectivos , Fatores de Risco , Redução de Peso/genética
8.
BMB Rep ; 42(6): 380-6, 2009 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-19558798

RESUMO

In neurodegenerative diseases, such as Alzheimer's and Parkinson's, microglial cell activation is thought to contribute to CNS injury by producing neurotoxic compounds. Prothrombin and kringle-2 increase levels of NO and the mRNA expression of iNOS, IL-1beta, and TNF-alpha in microglial cells. In contrast, the human prothrombin kringle-2 derived peptide NSA9 inhibits NO release and the production of pro-inflammatory cytokines such as IL-1beta, TNF-alpha, and IL-6 in LPS-activated EOC2 microglia. In this study, we investigated the anti-inflammatory effects of NSA9 in human prothrombin- and kringle-2-stimulated EOC2 microglia. Treatment with 20-100 muM of NSA9 attenuated both prothrombin- and kringle-2-induced microglial activation. NO production induced by MAPKs and NF-kappaB was similarly reduced by inhibitors of ERK (PD98059), p38 (SB203580), NF-kappaB (N-acetylcysteine), and NSA9. These results suggest that NSA9 acts independently as an inhibitor of microglial activation and that its effects in EOC2 microglia are not influenced by the presence of kringle-2.


Assuntos
Linhagem Celular , Microglia/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Protrombina/química , Protrombina/farmacologia , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Kringles/fisiologia , Microglia/metabolismo , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Fagocitose/efeitos dos fármacos , Protrombina/fisiologia
9.
J Alzheimers Dis ; 16(2): 421-31, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19221431

RESUMO

Prion diseases are a group of fatal neurodegenerative disorders characterized by the accumulation of a misfolded form (PrP(Sc)) of the cellular prion protein (PrP(C)) in the brains of affected individuals. The conversion of PrP(C) to PrP(Sc) is thought to involve a change in protein conformation from a normal, primarily alpha-helical structure into a beta-sheet conformer. Few proteins have been identified that differentially interact with the two forms of PrP. It has been reported that plasminogen binds to PrP(Sc) from a variety of prion phenotypes. We have examined potential motifs within the kringle region that may be responsible for binding to PrP. We synthesized 12-15-mer peptides that contain small, repetitive stretches of amino acid residues found within the kringle domains of plasminogen. These synthetic peptides were found to capture PrP(Sc) from the brain homogenates of bovine spongiform encephalopathy affected cattle, chronic wasting disease affected elk, experimental scrapie of hamsters and that of subjects affected by Creutzfeldt-Jakob disease, without binding to PrP(C) in unaffected controls. Therefore, we have identified critical peptide motifs that may be important for protein-protein interactions in prion disease pathogenesis. The ability of these synthetic peptides to bind preferentially to PrP(Sc) suggests a potential application in the diagnosis of prion diseases.


Assuntos
Encéfalo/metabolismo , Kringles/fisiologia , Fragmentos de Peptídeos/química , Doenças Priônicas/patologia , Príons/metabolismo , Animais , Bovinos , Síndrome de Creutzfeldt-Jakob/metabolismo , Síndrome de Creutzfeldt-Jakob/patologia , Cervos , Humanos , Imunoensaio/métodos , Mutação , Fragmentos de Peptídeos/farmacologia , Plasminogênio/química , Príons/química , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Conformação Proteica/efeitos dos fármacos , Doença de Emaciação Crônica/metabolismo , Doença de Emaciação Crônica/patologia
10.
Mol Cell Biochem ; 322(1-2): 1-13, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18979187

RESUMO

Vascular reorganization in wound healing is a complex process, which involves coagulation, endothelial cell proliferation and migration, basement membrane regeneration, and fibrinolysis. During this healing process, the hemostatic system and the angiogenic system are intimately interconnected. To elucidate the contribution of plasminogen in the process of wound healing, we have established a perfusion cell culture system. Using this novel cell culture system, we found that addition of plasminogen in the perfusion medium allowed the "scratch-wounded" endothelial cells to recover completely, while mini-plasminogen only affected the migration but not the proliferation of the endothelial cells. In the process of recovery with the addition of plasminogen, significant plasmin activity could only be detected when the growth of the endothelial cells have almost reached confluence. This finding indicates that wound healing is triggered and promoted during the absence of the proteolytic activity of plasmin. In addition, we could not detect any matrix metalloproteinase activity in the perfusion culture medium throughout the whole culture period. However, we did found that the circulating medium collected from the perfusion system at the early phase of the healing process has stimulatory activity on the growth of endothelial cells, but the proliferative activity decreased back to the basal level when the cells reached confluence. Thus, by using the perfusion cell culture system, we found that proliferation of endothelial cells is regulated by plasminogen and the wound healing process is controlled by a temporal interaction between the endothelial cells and plasminogen.


Assuntos
Técnicas de Cultura de Células , Plasminogênio/metabolismo , Cicatrização/fisiologia , Animais , Bovinos , Proliferação de Células , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Fibrinolisina/metabolismo , Kringles/fisiologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo
11.
Beijing Da Xue Xue Bao Yi Xue Ban ; 40(6): 572-7, 2008 Dec 18.
Artigo em Chinês | MEDLINE | ID: mdl-19088825

RESUMO

OBJECTIVE: To find novel isoform of PIK3IP1 and analyze their effects on cell viability, subcellular localization, and expression profile in cell lines. METHODS: RT-PCR was used to clone PIK3IP1 and its novel splicing isoform PIK3IP1-v1 from multi-tissue cDNA pool. By cell-based assays, we studied how PIK3IP1 and PIK3IP1-v1 affected the activity of Renila luciferase and morphological change in the HEK 293T cells. Furthermore, flow cytometry experiment was used to validate that overexpression of both splicing isoforms could induce cell apoptosis. Bioinformatics analysis was used to identify structural characteristics of these two splicing isoforms. By fluorescence microscopy assay, we identified their subcellular localization. RT-PCR was used to detect the expression of PIK3IP1 in the cell lines. RESULTS: PIK3IP1 and its novel splicing isoform PIK3IP1-v1 were cloned and constructed into the pcDNA-and pEGFP-expression plasmids. They both had signal peptide and transmembrane domain. Nevertheless, PIK3IP1-v1 was in absence of an extracellular Kringle domain. They could inhibit the activity of Renila luciferase and induce cell apoptosis. Simultaneously, both splicing isoforms are validated with subcellular localization on cell membrane and lowly expressed in many cell lines. CONCLUSION: PIK3IP1-v1 is a novel splicing isoform of PIK3IP1. Both of them are located on cell membrane and can induce cell apoptosis.


Assuntos
Apoptose/fisiologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Processamento Alternativo , Linhagem Celular , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Kringles/fisiologia , Ligação Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Subunidades Proteicas
13.
J Mol Med (Berl) ; 86(10): 1127-38, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18545979

RESUMO

Targeted therapy is a new generation of therapeutics, where two critical factors are involved. One is the particular molecular target, and the other is the specific target-binding drug. In this work, the fibrin, a main component of plasma clot at wound sites, was used as the target for human bFGF, aiming to improve therapeutic neovascularization and wound repair. To endow bFGF with fibrin-targeting ability, a fibrin-binding peptide Kringle1 (K1), derived from human plasminogen, was fused to human bFGF. The recombinant K1bFGF showed high fibrin and plasma-clot-binding ability. When applied to the wound sites with plasma clots, K1bFGF induced robust neovascularization and improved wound healing. To extend the application of K1bFGF to other cases where no plasma clots exist, we developed a fibrin-scaffold/K1bFGF system. This system could induce localized neovascularization by delivery of K1bFGF in a sustained and site-targeting manner, and provide a microenvironment promoting cell growth and tissue regeneration. In summary, we successfully used the pathologic environment fibrin clot as the target for bFGF, and based on which bFGF was designed into a targeting agent by introduction of a fibrin-binding peptide. This provides a potential approach to improve therapeutic neovascularization and wound repair.


Assuntos
Fibrina/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Kringles/fisiologia , Neovascularização Fisiológica/fisiologia , Cicatrização/fisiologia , Animais , Western Blotting , Ensaio de Imunoadsorção Enzimática , Fibrina/ultraestrutura , Fator 2 de Crescimento de Fibroblastos/genética , Humanos , Kringles/genética , Masculino , Microscopia Eletrônica de Varredura , Neovascularização Fisiológica/genética , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Cicatrização/genética
14.
Biochem Biophys Res Commun ; 369(2): 635-40, 2008 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-18294956

RESUMO

Angiostatin, a potent inhibitor of angiogenesis, is derived from the fibrinolytic proenzyme, plasminogen, by enzymatic processing. Plasminogen N-terminal activation peptide (PAP) is one of the products concomitantly released aside from angiostatin (kringles 1-4) and mini-plasminogen (kringle 5 plus the catalytic domain) when plasminogen is processed. To determine whether PAP alone or together with the angiostatin-related peptides derived from the processing of plasminogen modulate the proliferation and motility of endothelial cells, we have generated a recombinant PAP and used it to study its effects on endothelial cells in the presence and absence of the angiostatin-related peptides. Our results showed that PAP alone slightly increased the migration but not the proliferation of endothelial cells. However, in the presence of the angiostatin-related peptides, PAP attenuated the inhibitory activity of the angiostatin-related peptides on the proliferation and migration of endothelial cells. The inhibitory effect of PAP on the angiostatin-related peptides could be due to its binding to the kringle domains of the latter peptides.


Assuntos
Proteínas Angiostáticas/administração & dosagem , Proliferação de Células/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Plasminogênio/administração & dosagem , Células Cultivadas , Interações Medicamentosas , Células Endoteliais/citologia , Humanos , Kringles/fisiologia
15.
Biol Chem ; 389(2): 135-41, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18163888

RESUMO

Abstract Angiogenesis plays an important role in normal physiology of blood vessel growth, but can contribute to the pathogenesis of diseases, such as cancer. A new anti-angiogenic recombinant kringle protein, composed of the fused domains of human apolipoprotein(a) carboxyl-terminal kringle IV-10 and kringle V, was expressed in Pichia pastoris and human colorectal carcinoma (HCT 116) cells to investigate its influence on angiogenesis and tumor growth. The mature recombinant protein exhibited the characteristic features of kringle-containing proteins (glycosylation and disulfide bond formation) and, when added to cultures of human umbilical vein endothelial cell, resulted in a 31% decrease in proliferation relative to untreated controls (p<0.05). The neo-angiogenesis was diminished by 63% in chick embryos treated with 10 mug recombinant protein compared with 7% for phosphate buffer solution-treated embryos (p<0.01). Transfection of a kringle IV-10-kringle V fusion protein construct into HCT 116 cells decreased tumorigenesis and inhibited tumor growth in vivo without affecting tumor cell proliferation. HCT 116 cells that expressed recombinant protein displayed a much lower relative growth ratio of 8% (p<0.01) against the control tumor cells. From these results, we conclude that human apolipoprotein(a) carboxyl-terminal kringle IV-10-kringle V fusion protein is an effective inhibitor of angiogenesis and angiogenesis-dependent tumor growth.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Antineoplásicos/uso terapêutico , Apolipoproteínas A/química , Kringles/genética , Neoplasias Experimentais/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Inibidores da Angiogênese/genética , Inibidores da Angiogênese/metabolismo , Animais , Apolipoproteínas A/metabolismo , Apolipoproteínas A/fisiologia , Linhagem Celular Tumoral , Embrião de Galinha , Clonagem Molecular , Neoplasias Colorretais/patologia , Humanos , Kringles/fisiologia , Camundongos , Camundongos Nus , Neoplasias Experimentais/patologia , Neovascularização Patológica/prevenção & controle , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/uso terapêutico , Transplante Heterólogo
16.
Cancer Res ; 68(2): 404-14, 2008 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-18199534

RESUMO

The kringle 1 domain of human hepatocyte growth factor (HGFK1) was previously shown to inhibit bovine aortic endothelial cell proliferation, suggesting that it might be an antiangiogenic molecule. Here, we evaluated the in vivo efficacy of a recombinant adenoassociated virus carrying HGFK1 (rAAV-HGFK1) for the treatment of hepatocellular carcinoma (HCC) in a rat orthotopic HCC model and explored its molecular mechanisms in vitro in both endothelial and tumor cells. We first showed that rAAV-HGFK1 treatment significantly prolonged the survival time of rats transplanted with tumor cells. Treatment with rAAV-HGFK1 inhibited tumor growth, decreased tumor microvessel density, and completely prevented intrahepatic, lung, and peritoneal metastasis in this in vivo model. In vitro, rAAV-HGFK1 exhibited both antiangiogenic and antitumor cell effects, inhibiting the proliferation of both murine microvascular endothelial cells (MEC) and tumor cells, and inducing apoptosis and G(0)-G(1) phase arrest in these cells. To our surprise, rAAV-HGFK1 did not act through the hepatocyte growth factor/hepatocyte growth factor receptor pathway. Instead, it worked mainly through epidermal growth factor (EGF)/epidermal growth factor receptor (EGFR) signaling, with more minor contributions from vascular endothelial growth factor/vascular endothelial growth factor receptor and beta fibroblast growth factor (bFGF)/beta fibroblast growth factor receptor (bFGFR) signaling. In both MECs and tumor cells, rAAV-HGFK1 acted through two pathways downstream of EGFR, namely inhibition of extracellular signal-regulated kinase activation and stimulation of p38 mitogen-activated protein kinase/c-Jun-NH(2)-kinase activation. These results suggest for the first time that HGFK1 exerts both antiangiogenic and antitumor cell activities mainly through EGF/EGFR signaling, and may thus be considered as a novel therapeutic strategy for the treatment of HCC.


Assuntos
Carcinoma Hepatocelular/terapia , Terapia Genética , Fator de Crescimento de Hepatócito/química , Fator de Crescimento de Hepatócito/genética , Kringles/genética , Neoplasias Hepáticas Experimentais/terapia , Inibidores da Angiogênese/genética , Inibidores da Angiogênese/uso terapêutico , Animais , Antineoplásicos/metabolismo , Antineoplásicos/uso terapêutico , Apoptose/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/mortalidade , Carcinoma Hepatocelular/patologia , Ciclo Celular/genética , Proliferação de Células , Células Cultivadas , Dependovirus/genética , Receptores ErbB/antagonistas & inibidores , Fator de Crescimento de Hepatócito/uso terapêutico , Humanos , Kringles/fisiologia , Neoplasias Hepáticas Experimentais/genética , Neoplasias Hepáticas Experimentais/mortalidade , Neoplasias Hepáticas Experimentais/patologia , Masculino , Metástase Neoplásica , Ratos , Ratos Endogâmicos BUF , Receptores de Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Análise de Sobrevida
17.
Biochemistry ; 46(30): 8879-87, 2007 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-17616171

RESUMO

Streptokinase may be less effective at saving lives in patients with heart attacks because it explosively generates plasmin in the bloodstream at sites distant from fibrin clots. We hypothesized that this rapid plasmin generation is due to SK's singular capacity to nonproteolytically generate the active protease SK x Pg*, and we examined whether the kringle domains regulate this process. An SK mutant lacking Ile-1 (deltaIle1-SK) does not form SK x Pg*, although it will form complexes with plasmin that can activate plasminogen. When compared to SK, deltaIle1-SK diminished the generation of plasmin in plasma by more than 30-fold, demonstrating that the formation of SK x Pg* plays an important role in SK activity in the blood. The rate of SK x Pg* formation (measured by an active site titrant) was much slower in Glu-Pg, which contains five kringle domains, than in Pg forms containing one kringle (mini-Pg) or no kringles (micro-Pg). In a similar manner, Streptococcus uberis Pg activator (SUPA), an SK-like molecule, generated SUPA x Pg* much slower with bovine Pg than bovine micro-Pg. The velocity of SK x Pg* formation was regulated by agents that influence the conformation of Pg through interactions with the kringle domains. Chloride ions, which maintain the compact Pg conformation, hindered SK x Pg* formation. In contrast, epsilon-aminocaproic acid, fibrin, and fibrinogen, which induce an extended Pg conformation, accelerated the formation of SK x Pg*. In summary, the explosive generation of plasmin in blood or plasma, which diminishes SK's therapeutic effects, is attributable to the formation of SK x Pg*, and this process is governed by kringle domains.


Assuntos
Ativação Enzimática/fisiologia , Plasminogênio/química , Plasminogênio/metabolismo , Regulação Alostérica , Ácido Aminocaproico/química , Animais , Sítios de Ligação/fisiologia , Domínio Catalítico/fisiologia , Bovinos , Fibrina/química , Fibrina/metabolismo , Fibrinogênio/química , Fibrinogênio/metabolismo , Fibrinolisina/metabolismo , Humanos , Cinética , Kringles/fisiologia , Metaloendopeptidases/química , Metaloendopeptidases/metabolismo , Fragmentos de Peptídeos/metabolismo , Ativadores de Plasminogênio/química , Ativadores de Plasminogênio/metabolismo , Ligação Proteica , Estreptoquinase/química , Estreptoquinase/metabolismo , Relação Estrutura-Atividade , Ativador de Plasminogênio Tecidual/metabolismo
18.
FASEB J ; 21(14): 3928-36, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17622568

RESUMO

In light of the involvement of tumor-associated macrophages (TAM) in the promotion of tumor growth and metastasis, strategies to prevent TAM recruitment within the tumor microenvironment are currently under investigation. The recent observation that angiostatin reduces macrophage infiltration in an atherosclerosis model prompted our laboratory to further explore the use of human plasminogen angiostatin (hK1-3) protein as a macrophage modulatory agent. We demonstrate that hK1-3 blocks migration of murine peritoneal macrophages (91% decrease, P<0.00005) and human monocytes (85% decrease, P<0.05) in vitro. Cell viability of hK1-3-treated cells is not affected, as determined by fluorochrome-labeled inhibitors of caspase-propidium iodide (FLICA/PI) flow cytometry analysis. Furthermore, confocal microscopy of phalloidin-stained cells reveals that hK1-3 leads to disruption of actin filopodia/lamellipodia in human monocytes and induces distinct podosome accumulation in mature differentiated macrophages. Paradoxically, we observed a 3.5-fold increase in secretion and a 3- to 5.5-fold increase in gelatinolytic activity of macrophage-produced matrix metalloproteinase-9, which we suggest is a cellular response to compensate for the dominant static effect of hK1-3 on actin. We also demonstrate that hK1-3 induces the phosphorylation of extracellular signal-regulated kinase (ERK1/2) in human monocytes. hK1-3-mediated macrophage immobilization has the potential to be exploited therapeutically in pathological conditions associated with cellular hypoxia, such as cancer and atherosclerosis.


Assuntos
Actinas/antagonistas & inibidores , Angiostatinas/fisiologia , Inibição de Migração Celular/fisiologia , Citoesqueleto/metabolismo , Macrófagos Peritoneais/metabolismo , Monócitos/metabolismo , Actinas/metabolismo , Animais , Células Cultivadas , Citoesqueleto/patologia , Humanos , Kringles/fisiologia , Macrófagos Peritoneais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/patologia , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Fragmentos de Peptídeos/fisiologia
19.
J Surg Res ; 141(1): 83-90, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17574041

RESUMO

BACKGROUND: Urokinase plasminogen activator (uPA) is involved in vessel remodeling and mediates smooth muscle cell migration. Migration in response to uPA is dependent on both the growth factor binding domain at the aminoterminal end and the kringle (K) domain of the molecule. uPA is readily degraded in vivo into these constitutive domains. The aim of this study was to examine cell signaling during the migration of smooth muscle cell in response to the kringle domain of urokinase. MATERIALS AND METHODS: Murine arterial smooth muscle cells were cultured in vitro. Migration assays were performed in the presence of K with and without the plasmin inhibitors (aprotinin and -aminocaproic acid), the Galphai inhibitor Pertussis toxin, the MMP inhibitor (GM6001), the PI3-K inhibitors, Wortmannin and LY294002, and the MAPK inhibitors PD98089 (MEK1 inhibitor) and SB203580 (p38(MAPK) inhibitor). Western blotting was performed for ERK 1/2 and p38(MAPK) phosphorylation after stimulation with K in the presence and absence of the inhibitors. Statistics were analyzed by one-way ANOVA (n = 6). RESULTS: The kringle domain (K) induced a plasmin-independent, MMP-dependent increase in cell migration (2-fold, P < 0.05) compared to control. This migratory response to K was Galphai mediated and dependent on both ERK 1/2 and p38(MAPK) activation. K induced time-dependent increases in the phosphorylation of ERK 1/2 (3-fold, P < 0.05) and p38(MAPK) (3-fold, P < 0.05). Activation of p38(MAPK) and ERK 1/2 was completely inhibited by the PI3-K inhibitors. We explored a potential role for the epidermal growth factor receptor (EGFR). K induced EGFR phosphorylation and the presence of AG1478, the EGFR inhibitor, inhibited both cell migration and akt activation in response to K. CONCLUSION: Kringle domain of uPA induces smooth muscle cell migration through a G-protein-coupled PI3-K-dependent process involving both ERK 1/2 and p38(MAPK) and is mediated in part through EGFR. Defining the differences in response to key molecular domains of uPA is vital to understand its role in vessel remodeling.


Assuntos
Movimento Celular/fisiologia , Kringles/fisiologia , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiologia , Ativador de Plasminogênio Tipo Uroquinase/fisiologia , Animais , Células Cultivadas , Receptores ErbB/fisiologia , Proteínas de Ligação ao GTP/fisiologia , Camundongos , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
20.
J Biotechnol ; 126(4): 562-7, 2006 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16797763

RESUMO

The GAL1 gene encoding galactokinase was disrupted in a recombinant Saccharomyces cerevisiae strain in which production of LK8 protein, a kringle fragment of human apolipoprotein, is under the control of GAL1 promoter. Null mutation of the HXK2 gene was introduced further in the gal1Delta strain to relieve glucose repression. A pattern for LK8 expression was compared for the two recombinant S. cerevisiae systems in continuous and fed-batch cultivations. A critical dilution rate in continuous cultivation that repressed LK8 expression was significantly higher for the gal1Deltahxk2Delta strain than that for the gal1Delta strain to sustain the LK8 production even at high glucose consumption rate. Expressed LK8 for the gal1Delta strain was not detectable when the dilution rate exceeded 0.05 h(-1). Maximum LK8 concentration of 57 mgl(-1) was obtained in glucose-limited fed-batch cultivation of the gal1Deltahxk2Delta strain, corresponding to a 13.8-fold enhancement compared with the gal1Delta strain grown under the same conditions.


Assuntos
Glucose/metabolismo , Hexoquinase/genética , Kringles/fisiologia , Saccharomyces cerevisiae/metabolismo , Reatores Biológicos/microbiologia , Deleção de Genes , Hexoquinase/metabolismo , Humanos , Kringles/genética , Regiões Promotoras Genéticas , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crescimento & desenvolvimento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...