Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 467
Filtrar
1.
Int J Mol Sci ; 25(13)2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-39000111

RESUMEN

A new family of antifibrinolytic drugs has been recently discovered, combining a triazole moiety, an oxadiazolone, and a terminal amine. Two of the molecules of this family have shown activity that is greater than or similar to that of tranexamic acid (TXA), the current antifibrinolytic gold standard, which has been associated with several side effects and whose use is limited in patients with renal impairment. The aim of this work was to thoroughly examine the mechanism of action of the two ideal candidates of the 1,2,3-triazole family and compare them with TXA, to identify an antifibrinolytic alternative active at lower dosages. Specifically, the antifibrinolytic activity of the two compounds (1 and 5) and TXA was assessed in fibrinolytic isolated systems and in whole blood. Results revealed that despite having an activity pathway comparable to that of TXA, both compounds showed greater activity in blood. These differences could be attributed to a more stable ligand-target binding to the pocket of plasminogen for compounds 1 and 5, as suggested by molecular dynamic simulations. This work presents further evidence of the antifibrinolytic activity of the two best candidates of the 1,2,3-triazole family and paves the way for incorporating these molecules as new antifibrinolytic therapies.


Asunto(s)
Antifibrinolíticos , Ácido Tranexámico , Triazoles , Triazoles/química , Triazoles/farmacología , Antifibrinolíticos/farmacología , Antifibrinolíticos/química , Humanos , Ácido Tranexámico/farmacología , Ácido Tranexámico/química , Simulación de Dinámica Molecular , Plasminógeno/metabolismo , Plasminógeno/química , Fibrinólisis/efectos de los fármacos
2.
ACS Appl Mater Interfaces ; 16(28): 35874-35886, 2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-38954798

RESUMEN

To effectively solve the problem of significant loss of transplanted cells caused by thrombosis during cell transplantation, this study simulates the human fibrinolytic system and combines metabolic oligosaccharide engineering with strain-promoted azide-alkyne cycloaddition (SPAAC) click chemistry to construct a cell surface with fibrinolytic activity. First, a copolymer (POL) of oligoethylene glycol methacrylate (OEGMA) and 6-amino-2-(2-methylamido)hexanoic acid (Lys) was synthesized by reversible addition-fragmentation chain transfer (RAFT) copolymerization, and the dibenzocyclooctyne (DBCO) functional group was introduced into the side chain of the copolymer through an active ester reaction, resulting in a functionalized copolymer DBCO-PEG4-POL with ε-lysine ligands. Then, azide functional groups were introduced onto the surface of HeLa model cells through metabolic oligosaccharide engineering, and DBCO-PEG4-POL was further specifically modified onto the surface of HeLa cells via the SPAAC "click" reaction. In vitro investigations revealed that compared with unmodified HeLa cells, modified cells not only resist the adsorption of nonspecific proteins such as fibrinogen and human serum albumin but also selectively bind to plasminogen in plasma while maintaining good cell viability and proliferative activity. More importantly, upon the activation of adsorbed plasminogen into plasmin, the modified cells exhibited remarkable fibrinolytic activity and were capable of promptly dissolving the primary thrombus formed on their surfaces. This research not only provides a novel approach for constructing transplantable cells with fibrinolytic activity but also offers a new perspective for effectively addressing the significant loss of transplanted cells caused by thrombosis.


Asunto(s)
Química Clic , Reacción de Cicloadición , Fibrinólisis , Oligosacáridos , Humanos , Células HeLa , Oligosacáridos/química , Fibrinólisis/efectos de los fármacos , Ingeniería Metabólica , Azidas/química , Polietilenglicoles/química , Metacrilatos/química , Alquinos/química , Animales , Supervivencia Celular/efectos de los fármacos , Plasminógeno/química , Plasminógeno/metabolismo , Propiedades de Superficie
3.
Nature ; 629(8013): 945-950, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38720069

RESUMEN

Lipoprotein(a) (Lp(a)), an independent, causal cardiovascular risk factor, is a lipoprotein particle that is formed by the interaction of a low-density lipoprotein (LDL) particle and apolipoprotein(a) (apo(a))1,2. Apo(a) first binds to lysine residues of apolipoprotein B-100 (apoB-100) on LDL through the Kringle IV (KIV) 7 and 8 domains, before a disulfide bond forms between apo(a) and apoB-100 to create Lp(a) (refs. 3-7). Here we show that the first step of Lp(a) formation can be inhibited through small-molecule interactions with apo(a) KIV7-8. We identify compounds that bind to apo(a) KIV7-8, and, through chemical optimization and further application of multivalency, we create compounds with subnanomolar potency that inhibit the formation of Lp(a). Oral doses of prototype compounds and a potent, multivalent disruptor, LY3473329 (muvalaplin), reduced the levels of Lp(a) in transgenic mice and in cynomolgus monkeys. Although multivalent molecules bind to the Kringle domains of rat plasminogen and reduce plasmin activity, species-selective differences in plasminogen sequences suggest that inhibitor molecules will reduce the levels of Lp(a), but not those of plasminogen, in humans. These data support the clinical development of LY3473329-which is already in phase 2 studies-as a potent and specific orally administered agent for reducing the levels of Lp(a).


Asunto(s)
Descubrimiento de Drogas , Lipoproteína(a) , Macaca fascicularis , Animales , Femenino , Humanos , Masculino , Ratones , Administración Oral , Kringles , Lipoproteína(a)/antagonistas & inhibidores , Lipoproteína(a)/sangre , Lipoproteína(a)/química , Lipoproteína(a)/metabolismo , Ratones Transgénicos , Bibliotecas de Moléculas Pequeñas/farmacología , Bibliotecas de Moléculas Pequeñas/química , Plasminógeno/química , Plasminógeno/metabolismo , Especificidad de la Especie , Ensayos Clínicos Fase II como Asunto , Apolipoproteínas A/química , Apolipoproteínas A/metabolismo
4.
Int J Biol Macromol ; 270(Pt 1): 132356, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38754659

RESUMEN

The potent angiogenesis inhibitor known as human plasminogen Kringle 5 has shown promise in the treatment of vascular disorders and malignancies. The study aimed to investigate the recognition and interaction between Kringle 5 and the A2M domain of human complement component C5 using bio-specific methodologies and molecular dynamics (MD) simulation. Initially, the specific interaction between Kringle 5 and A2M was confirmed and characterized through Ligand Blot and ELISA, yielding the dissociation constant (Kd) of 1.70 × 10-7 mol/L. Then, Kringle 5 showcased a dose-dependent inhibition of the production of C5a in lung cancer A549 cells, consequently impeding their proliferation and migration. Following the utilization of frontal affinity chromatography (FAC), it was revealed that there exists a singular binding site with the binding constant (Ka) of 3.79 × 105 L/mol. Following the implementation of homology modeling and MD optimization, the detailed results indicate that only a specific segment of the N-terminal structure of the A2M molecule engages in interaction with Kringle 5 throughout the binding process and the principal driving forces encompass electrostatic force, hydrogen bonding, and van der Waals force. In conclusion, the A2M domain of human complement C5 emerges as a plausible binding target for Kringle 5 in vivo.


Asunto(s)
Simulación de Dinámica Molecular , Plasminógeno , Unión Proteica , Humanos , Plasminógeno/química , Plasminógeno/metabolismo , Sitios de Unión , Complemento C5a/química , Complemento C5a/metabolismo , Células A549 , Dominios Proteicos , Proliferación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Fragmentos de Péptidos
5.
J Biomol Struct Dyn ; 42(6): 3204-3222, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37216286

RESUMEN

The zymogen protease Plasminogen (Plg) and its active form plasmin (Plm) carry out important functions in the blood clot disintegration (breakdown of fibrin fibers) process. Inhibition of plasmin effectively reduces fibrinolysis to circumvent heavy bleeding. Currently, available Plm inhibitor tranexamic acid (TXA) used for treating severe hemorrhages is associated with an increased incidence of seizures which in turn were traced to gamma-aminobutyric acid antagonistic activity (GABAa) in addition to having multiple side effects. Fibrinolysis can be suppressed by targeting the three important protein domains: the kringle-2 domain of tissue plasminogen activator, the kringle-1 domain of plasminogen, and the serine protease domain of plasminogen. In the present study, one million molecules were screened from the ZINC database. These ligands were docked to their respective protein targets using Autodock Vina, Schrödinger Glide, and ParDOCK/BAPPL+. Thereafter, the drug-likeness properties of the ligands were evaluated using Discovery Studio 3.5. Subsequently, we subjected the protein-ligand complexes to molecular dynamics simulation of 200 ns in GROMACS. The identified ligands P76(ZINC09970930), C97(ZINC14888376), and U97(ZINC11839443) for each protein target are found to impart higher stability and greater compactness to the protein-ligand complexes. Principal component analysis (PCA) implicates, that the identified ligands occupy smaller phase space, form stable clusters, and provide greater rigidity to the protein-ligand complexes. Molecular Mechanics Poisson-Boltzmann Surface Area (MMPBSA) analysis reveals that P76, C97, and U97 exhibit better binding free energy (ΔG) when compared to that of the standard ligands. Thus, our findings can be useful for the development of promising anti-fibrinolytic agents.Communicated by Ramaswamy H. Sarma.


Asunto(s)
Plasminógeno , Activador de Tejido Plasminógeno , Plasminógeno/química , Plasminógeno/metabolismo , Plasminógeno/farmacología , Activador de Tejido Plasminógeno/química , Activador de Tejido Plasminógeno/metabolismo , Activador de Tejido Plasminógeno/farmacología , Fibrinolisina/metabolismo , Ligandos , Fibrinólisis
6.
Arterioscler Thromb Vasc Biol ; 43(10): 1851-1866, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37589135

RESUMEN

BACKGROUND: High levels of Lp(a) (lipoprotein(a)) are associated with multiple forms of cardiovascular disease. Lp(a) consists of an apoB100-containing particle attached to the plasminogen homologue apo(a). The pathways for Lp(a) clearance are not well understood. We previously discovered that the plasminogen receptor PlgRKT (plasminogen receptor with a C-terminal lysine) promoted Lp(a) uptake in liver cells. Here, we aimed to further define the role of PlgRKT and to investigate the role of 2 other plasminogen receptors, annexin A2 and S100A10 (S100 calcium-binding protein A10) in the endocytosis of Lp(a). METHODS: Human hepatocellular carcinoma (HepG2) cells and haploid human fibroblast-like (HAP1) cells were used for overexpression and knockout of plasminogen receptors. The uptake of Lp(a), LDL (low-density lipoprotein), apo(a), and endocytic cargos was visualized and quantified by confocal microscopy and Western blotting. RESULTS: The uptake of both Lp(a) and apo(a), but not LDL, was significantly increased in HepG2 and HAP1 cells overexpressing PlgRKT, annexin A2, or S100A10. Conversely, Lp(a) and apo(a), but not LDL, uptake was significantly reduced in HAP1 cells in which PlgRKT and S100A10 were knocked out. Surface binding studies in HepG2 cells showed that overexpression of PlgRKT, but not annexin A2 or S100A10, increased Lp(a) and apo(a) plasma membrane binding. Annexin A2 and S100A10, on the other hand, appeared to regulate macropinocytosis with both proteins significantly increasing the uptake of the macropinocytosis marker dextran when overexpressed in HepG2 and HAP1 cells and knockout of S100A10 significantly reducing dextran uptake. Bringing these observations together, we tested the effect of a PI3K (phosphoinositide-3-kinase) inhibitor, known to inhibit macropinocytosis, on Lp(a) uptake. Results showed a concentration-dependent reduction confirming that Lp(a) uptake was indeed mediated by macropinocytosis. CONCLUSIONS: These findings uncover a novel pathway for Lp(a) endocytosis involving multiple plasminogen receptors that enhance surface binding and stimulate macropinocytosis of Lp(a). Although the findings were produced in cell culture models that have limitations, they could have clinical relevance since drugs that inhibit macropinocytosis are in clinical use, that is, the PI3K inhibitors for cancer therapy and some antidepressant compounds.


Asunto(s)
Anexina A2 , Plasminógeno , Humanos , Plasminógeno/química , Plasminógeno/metabolismo , Lipoproteína(a)/metabolismo , Anexina A2/genética , Dextranos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Portadoras , Apolipoproteínas A/metabolismo
7.
Arch Biochem Biophys ; 743: 109671, 2023 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-37336343

RESUMEN

Staphylokinase (SAK), a potent fibrin-specific plasminogen activator secreted by Staphylococcus aureus, carries a pair of lysine at the carboxy-terminus that play a key role in plasminogen activation. The underlaying mechanism by which C-terminal lysins of SAK modulate its function remains unknown. This study has been undertaken to unravel role of C-terminal lysins of SAK in plasminogen activation. While deletion of C-terminal lysins (Lys135, Lys136) drastically impaired plasminogen activation by SAK, addition of lysins enhanced its catalytic activity 2-2.5-fold. Circular dichroism analysis revealed that C-terminally modified mutants of SAK carry significant changes in their beta sheets and secondary structure. Structure models and RING (residue interaction network generation) studies indicated that the deletion of lysins has conferred extensive topological alterations in SAK, disrupting vital interactions at the interface of SAK.plasmin complex, thereby leading significant impairment in its functional activity. In contrast, addition of lysins at the C-terminus enhanced its conformational flexibility, creating a stronger coupling at the interface of SAK.plasmin complex and making it more efficient for plasminogen activation. Taken together, these studies provided new insights on the role of C-terminal lysins in establishment of precise intermolecular interactions of SAK with the plasmin for the optimal function of activator complex.


Asunto(s)
Fibrinolisina , Lisina , Fibrinolisina/química , Plasminógeno/química , Activadores Plasminogénicos/química
8.
Bioorg Chem ; 137: 106579, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37149949

RESUMEN

Plasminogen Kringle 5 is one of the most potent cytokines identified to inhibit the proliferation and migration of vascular endothelial cells. Herein, six aptamer candidates that specifically bind to Kringle 5 were generated by the systematic evolution of ligands by exponential enrichment (SELEX). After 10 rounds of screening against Kringle 5, a highly enriched ssDNA pool was sequenced and the representative aptamers were subjected to binding assays to evaluate their affinity and specificity. The preferred aptamer KG-4, which demonstrated a low dissociation constant (Kd) of âˆ¼ 432 nM and excellent selectivity for Kringle 5. A conserved "motif" of eight bases located at the stem-loop intersection, common to the aptamer, was further confirmed as the recognition element for binding with Kringle 5. The bulge formed by the motif and depression on the lysine binding site of Kringle 5 were both located at the binding interface, and the "induced fit" between their structures played a central role in the recognition process. Kringle 5 interacts KG-4 primarily through enthalpy-driven van der Waals forces and hydrogen bond. The key nucleotides A34 and C35 at motif on KG-4 and the positively charged amino acids in the loop 1 and loop 4 regions on Kringle 5 play a major role in the interaction. Furthermore, KG-4 dose-dependently reduced the proliferation inhibition of vascular endothelial cells by Kringle 5 and had a blocking effect on the function of Kringle 5 in inhibiting migration and promoting apoptosis of vascular endothelial cells in vitro. This study put a new light on protein-aptamer binding mechanism and may provide insight into the treatment of ischemic diseases by target depletion of Kringle 5.


Asunto(s)
Aptámeros de Nucleótidos , Células Endoteliales , Humanos , Células Endoteliales/metabolismo , Plasminógeno/química , Plasminógeno/metabolismo , Sitios de Unión , Aptámeros de Nucleótidos/química
9.
J Biomol Struct Dyn ; 41(24): 15023-15032, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36927470

RESUMEN

Tetranectin-plasminogen interaction plays a defining role in extracellular matrix degradation, enabling tumor cell invasion and metastasis. This interaction occurs via the carbohydrate recognition domain (CRD) and Kringle 4 domain of tetranectin and plasminogen, respectively, leading to activation of the plasminogen-cascade that triggers the proteolytic processes. Thus targeting this interaction represents an important strategy to suppress tumor cell migration and invasion. In this direction, we attempted to target the CRD of tetranectin to inhibit its interaction with the Kringle-4 domain of plasminogen using natural bioactive compounds. A cheminformatics pipeline for drug designing and screening was utilized to obtain lead compound(s) that exhibit conformationally and energetically viable CRD binding. Out of 206 compounds screened, diosgenin and scytonemin displayed the most favorable interactions with CRD. Short-term molecular dynamics simulations of 20 ns were employed to further study the conformational stability of both compounds with tetranectin CRD which reflected at the increased stability of diosgenin in the CRD binding pocket compared to scytonemin. Finally, an extended molecular dynamic simulation of 100 ns affirmed the robust and stable interaction of diosgenin with CRD. Furthermore, diosgenin was observed to exert a pronounced anti-proliferative effect on high tetranectin-expressing MDA-MB-231 breast cancer cells. The inhibitory effect of diosgenin on the tetranectin-plasminogen interaction was corroborated by the reduced migration and invasiveness of MDA-MB-231 cells under diosgenin treatment. Overall the study presents an alternate and safer approach to impede breast cancer metastasis and delineates the novel anti-metastatic activity of diosgenin.Communicated by Ramaswamy H. Sarma.


Asunto(s)
Neoplasias de la Mama , Diosgenina , Melanoma , Neoplasias Cutáneas , Humanos , Femenino , Plasminógeno/química , Plasminógeno/metabolismo , Proteínas Sanguíneas/química , Neoplasias de la Mama/tratamiento farmacológico
10.
J Thromb Haemost ; 21(6): 1630-1635, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36914052

RESUMEN

BACKGROUND: Streptococcus pyogenes (GAS) is a human bacterial pathogen that generates various mild to severe diseases. Worldwide, there are approximately 700 million cases of GAS infections per year. In some strains of GAS, the surface-resident M-protein, plasminogen-binding group A streptococcal M-protein (PAM), binds directly to human host plasminogen (hPg), where it is activated to plasmin through a mechanism involving a Pg/bacterial streptokinase (SK) complex as well as endogenous activators. Binding to Pg and its activation are dictated by selected sequences within the human host Pg protein, making it difficult to generate animal models to study this pathogen. OBJECTIVES: To develop a murine model for studying GAS infection by minimally modifying mouse Pg to enhance the affinity to bacterial PAM and sensitivity to GAS-derived SK. METHODS: We used a targeting vector that contained a mouse albumin-promoter and mouse/human hybrid plasminogen cDNA targeted to the Rosa26 locus. Characterization of the mouse strain consisted of both gross and histological techniques and determination of the effects of the modified Pg protein through surface plasmon resonance measurements, Pg activation analyses, and mouse survival post-GAS infection. RESULTS: We generated a mouse line expressing a chimeric Pg protein consisting of 2 amino acid substitutions in the heavy chain of Pg and a complete replacement of the mouse Pg light chain with the human Pg light chain. CONCLUSION: This protein demonstrated an enhanced affinity for bacterial PAM and sensitivity to activation by the Pg-SK complex, making the murine host susceptible to the pathogenic effects of GAS.


Asunto(s)
Streptococcus pyogenes , Estreptoquinasa , Animales , Ratones , Humanos , Estreptoquinasa/genética , Estreptoquinasa/química , Estreptoquinasa/metabolismo , Streptococcus pyogenes/química , Streptococcus pyogenes/genética , Streptococcus pyogenes/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas Portadoras/metabolismo , Plasminógeno/química , Unión Proteica
11.
Biochemistry ; 62(3): 735-746, 2023 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-36701429

RESUMEN

Cellular plasminogen (Pg) receptors (PgRs) are utilized to recruit Pg; stimulate its activation to the serine protease, plasmin (Pm); and sterically protect the surface Pm from inactivation by host inhibitors. One such PgR is the moonlighting enzyme, enolase, some of which leaves the cytoplasm and resides at the cell surface to potentially function as a PgR. Since microbes employ conscription of host Pg by PgRs as one virulence mechanism, we explored the structural basis of the ability of Streptococcus pyogenes enolase (Sen) to function in this manner. Employing single-particle cryo-electron microscopy (cryo-EM), recombinant Sen from S. pyogenes was modeled at 2.6 Å as a stable symmetrical doughnut-shaped homooctamer with point group 422 (D4) symmetry, with a monomeric subunit molecular weight of ∼49 kDa. Binding sites for hPg were reported in other studies to include an internal K252,255 and the COOH-terminal K434,435 residues of Sen. However, in native Sen, the latter are buried within the minor interfaces of the octamer and do not function as a Pg-binding epitope. Whereas Sen and hPg do not interact in solution, when Sen is bound to a surface, hPg interacts with Sen independently of K252,255,434,435. PgRs devoid of COOH-terminal lysine utilize lysine isosteres comprising a basic residue, "i", and an anionic residue at "i + 3" around one turn of an α-helix. We highlight a number of surface-exposed potential hPg-binding lysine isosteres and further conclude that while the octameric structure of Sen is critical for hPg binding, disruption of this octamer without dissociation exposes hPg-binding epitopes.


Asunto(s)
Proteínas Bacterianas , Plasminógeno , Plasminógeno/química , Plasminógeno/metabolismo , Proteínas Bacterianas/química , Streptococcus pyogenes , Microscopía por Crioelectrón , Unión Proteica , Fosfopiruvato Hidratasa/metabolismo , Lisina/química , Proteínas Portadoras/metabolismo , Serina Proteasas/metabolismo
12.
Curr Pharm Biotechnol ; 24(10): 1326-1334, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36424778

RESUMEN

BACKGROUND: Streptokinase, one of the most widely used thrombolytic medicines, is a favorable protein for site-specific PEGylation as it lacks any cysteine residues in its amino acid sequence; however, any changes in the protein's structure should be carefully planned to avoid undesired changes in its function. OBJECTIVES: This study aimed to design and produce novel di/tri-cysteine variants of streptokinase from previously developed cysteine analogues, Arg45, Glu263, and Arg319, as candidates for multiple site-specific PEGylation. METHODS: Using bioinformatics tools and site-directed mutagenesis, we incorporated concurrent mutations at Arg45, Glu263, and Arg319 (carried out in our previous study) to create di/tri-cysteine variants of streptokinase proteins (SK45-319cys, SK263-319cys, and SK45-263-319cys) and evaluated their kinetic activity parameters by a colorimetric method, using H-D-Val-Leu-Lys-pNA.2HCl (S2251) as substrate. RESULTS: Based on the kinetic results, SK263-319cys with 44% enzyme efficiency increment compared to wild-type SK was the superior protein in terms of activity; as well, SK45-319cys and SK45-263-319cys showed 17 and 22% activity enhancement, respectively. Docking of the mutant streptokinase proteins with µ-plasmin demonstrated that changes in intermolecular interactions caused by amino acid substitution could be the reason for activity difference. CONCLUSION: The novel mutant proteins created in this study exhibit remarkable biological activity and may be uniquely suitable for simultaneous PEGylation on two/three domains. As well, PEGylated derivates of these variants might prove to be more proficient proteins, compared to the singlecysteine analogs of streptokinase; because of their more surface coverage and increased molecular weight.


Asunto(s)
Cisteína , Estreptoquinasa , Estreptoquinasa/genética , Estreptoquinasa/metabolismo , Cisteína/genética , Plasminógeno/química , Plasminógeno/genética , Plasminógeno/metabolismo , Fibrinolíticos , Mutación
13.
Int J Biol Macromol ; 206: 64-73, 2022 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-35219777

RESUMEN

In this study, we examined for the first time the effect of the HOCl/OCl-- and H2O2-induced oxidation of Glu-plasminogen on damage to its primary structure and the biological activity of plasmin. The consolidated results obtained with the aid of MS/MS, electrophoresis, and colourimetry, demonstrated that none of the oxidised amino acid residues found in the proenzyme treated with 25 µM HOCl/OCl- or 100 µM H2O2 were functionally significant for plasminogen. However, the treatment of plasminogen with increasing concentrations of HOCl/OCl- from 25 µM to 100 µM or H2O2 from 100 µM to 300 µM promoted a partial loss in the activity of oxidised plasmin. Several methionine residues (Met57, Met182, Met385, Met404, Met585, and Met788) localized in different protein domains have been shown to serve as ROS traps, thus providing an efficient defense mechanism against oxidative stress. Oxidised Trp235, Trp417, Trp427, Trp761, and Tyr672 are most likely responsible for the reduced biological activity of Glu-plasminogen subjected to strong oxidation. The results of the present study, along with those of previous studies, indicate that the structure of Glu-plasminogen is adapted to oxidation to withstand oxidative stress induced by ROS.


Asunto(s)
Ácido Hipocloroso , Plasminógeno , Fibrinolisina , Peróxido de Hidrógeno , Ácido Hipocloroso/química , Peróxidos , Plasminógeno/química , Espectrometría de Masas en Tándem
14.
Microbiologyopen ; 10(6): e1252, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34964287

RESUMEN

The direct binding of human plasminogen (hPg), via its kringle-2 domain (K2hPg ), to streptococcal M-protein (PAM), largely contributes to the pathogenesis of Pattern D Group A Streptococcus pyogenes (GAS). However, the mechanism of complex formation is unknown. In a system consisting of a Class II PAM from Pattern D GAS isolate NS88.2 (PAMNS88.2 ), with one K2hPg binding a-repeat in its A-domain, we employed biophysical techniques to analyze the mechanism of the K2hPg /PAMNS88.2 interaction. We show that apo-PAMNS88.2 is a coiled-coil homodimer (M.Wt. ~80 kDa) at 4°C-25°C, and is monomeric (M.Wt. ~40 kDa) at 37°C, demonstrating a temperature-dependent dissociation of PAMNS88.2 over a narrow temperature range. PAMNS88.2 displayed a single tight binding site for K2hPg at 4°C, which progressively increased at 25°C through 37°C. We isolated the K2hPg /PAMNS88.2 complexes at 4°C, 25°C, and 37°C and found molecular weights of ~50 kDa at each temperature, corresponding to a 1:1 (m:m) K2hPg /PAMNS88.2  monomer complex. hPg activation experiments by streptokinase demonstrated that the hPg/PAMNS88.2  monomer complexes are fully functional. The data show that PAM dimers dissociate into functional monomers at physiological temperatures or when presented with the active hPg module (K2hPg ) showing that PAM is a functional monomer at 37°C.


Asunto(s)
Antígenos Bacterianos/química , Antígenos Bacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Kringles , Plasminógeno/química , Plasminógeno/metabolismo , Streptococcus pyogenes/metabolismo , Sitios de Unión , Humanos , Peso Molecular , Unión Proteica , Conformación Proteica en Hélice alfa , Multimerización de Proteína , Estreptoquinasa/metabolismo , Temperatura , Termodinámica
15.
Mar Drugs ; 19(6)2021 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-34071669

RESUMEN

Tetraselmis subcordiformis, a unicellular marine green alga, is used widely in aquaculture as an initial feeding for fish, bivalve mollusks, penaeid shrimp larvae, and rotifers because of its rich content of amino acids and fatty acids. A stable nuclear transformation system using the herbicide phosphinothricin (PPT) as a selective reagent was established previously. In this research, the recombinant expression in T. subcordiformis was investigated by particle bombardment with the rt-PA gene that encodes the recombinant human tissue-type plasminogen activator (Reteplase), which is a thrombolytic agent for acute myocardial infarction treatment. Transgenic algal strains were selected by their resistance to PPT, and expression of rt-PA was validated by PCR, Southern blotting, and Western blotting, and bioactivity of rt-PA was confirmed by the fibrin agarose plate assay for bioactivity. The results showed that rt-PA was integrated into the genome of T. subcordiformis, and the expression product was bioactive, indicating proper post-transcriptional modification of rt-PA in T. subcordiformis. This report contributes to efforts that take advantage of marine microalgae as cell factories to prepare recombinant drugs and in establishing a characteristic pathway of oral administration in aquaculture.


Asunto(s)
Chlorophyta/metabolismo , Fibrinolíticos/metabolismo , Microalgas/metabolismo , Activador de Tejido Plasminógeno/biosíntesis , Chlorophyta/genética , Microbiología Industrial , Microalgas/genética , Plasminógeno/química , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Activador de Tejido Plasminógeno/química , Activador de Tejido Plasminógeno/genética
16.
Int J Mol Sci ; 22(5)2021 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-33803235

RESUMEN

Fibrinolysis is an important process in hemostasis responsible for dissolving the clot during wound healing. Plasmin is a central enzyme in this process via its capacity to cleave fibrin. The kinetics of plasmin generation (PG) and inhibition during fibrinolysis have been poorly understood until the recent development of assays to quantify these metrics. The assessment of plasmin kinetics allows for the identification of fibrinolytic dysfunction and better understanding of the relationships between abnormal fibrin dissolution and disease pathogenesis. Additionally, direct measurement of the inhibition of PG by antifibrinolytic medications, such as tranexamic acid, can be a useful tool to assess the risks and effectiveness of antifibrinolytic therapy in hemorrhagic diseases. This review provides an overview of available PG assays to directly measure the kinetics of plasmin formation and inhibition in human and mouse plasmas and focuses on their applications in defining the role of plasmin in diseases, including angioedema, hemophilia, rare bleeding disorders, COVID-19, or diet-induced obesity. Moreover, this review introduces the PG assay as a promising clinical and research method to monitor antifibrinolytic medications and screen for genetic or acquired fibrinolytic disorders.


Asunto(s)
Análisis Químico de la Sangre/métodos , Enfermedad , Fibrinolisina/análisis , Fibrinolisina/metabolismo , Animales , Antifibrinolíticos/sangre , Fibrina/análisis , Fibrina/química , Fibrinolíticos/sangre , Humanos , Plasminógeno/análisis , Plasminógeno/química , Plasminógeno/metabolismo
17.
Sci Rep ; 11(1): 4964, 2021 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-33654183

RESUMEN

Borrelia miyamotoi, a relapsing fever spirochete transmitted by Ixodid ticks causes B. miyamotoi disease (BMD). To evade the human host´s immune response, relapsing fever borreliae, including B. miyamotoi, produce distinct variable major proteins. Here, we investigated Vsp1, Vlp15/16, and Vlp18 all of which are currently being evaluated as antigens for the serodiagnosis of BMD. Comparative analyses identified Vlp15/16 but not Vsp1 and Vlp18 as a plasminogen-interacting protein of B. miyamotoi. Furthermore, Vlp15/16 bound plasminogen in a dose-dependent fashion with high affinity. Binding of plasminogen to Vlp15/16 was significantly inhibited by the lysine analog tranexamic acid suggesting that the protein-protein interaction is mediated by lysine residues. By contrast, ionic strength did not have an effect on binding of plasminogen to Vlp15/16. Of relevance, plasminogen bound to the borrelial protein cleaved the chromogenic substrate S-2251 upon conversion by urokinase-type plasminogen activator (uPa), demonstrating it retained its physiological activity. Interestingly, further analyses revealed a complement inhibitory activity of Vlp15/16 and Vlp18 on the alternative pathway by a Factor H-independent mechanism. More importantly, both borrelial proteins protect serum sensitive Borrelia garinii cells from complement-mediated lysis suggesting multiple roles of these two variable major proteins in immune evasion of B. miyamotoi.


Asunto(s)
Proteínas Bacterianas , Borrelia , Proteínas del Sistema Complemento , Plasminógeno , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Borrelia/química , Borrelia/metabolismo , Proteínas del Sistema Complemento/química , Proteínas del Sistema Complemento/metabolismo , Humanos , Plasminógeno/química , Plasminógeno/metabolismo
18.
Blood ; 137(21): 2881-2889, 2021 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-33735914

RESUMEN

Plasminogen is an abundant plasma protein that exists in various zymogenic forms. Plasmin, the proteolytically active form of plasminogen, is known for its essential role in fibrinolysis. To date, therapeutic targeting of the fibrinolytic system has been for 2 purposes: to promote plasmin generation for thromboembolic conditions or to stop plasmin to reduce bleeding. However, plasmin and plasminogen serve other important functions, some of which are unrelated to fibrin removal. Indeed, for >40 years, the antifibrinolytic agent tranexamic acid has been administered for its serendipitously discovered skin-whitening properties. Plasmin also plays an important role in the removal of misfolded/aggregated proteins and can trigger other enzymatic cascades, including complement. In addition, plasminogen, via binding to one of its dozen cell surface receptors, can modulate cell behavior and further influence immune and inflammatory processes. Plasminogen administration itself has been reported to improve thrombolysis and to accelerate wound repair. Although many of these more recent findings have been derived from in vitro or animal studies, the use of antifibrinolytic agents to reduce bleeding in humans has revealed additional clinically relevant consequences, particularly in relation to reducing infection risk that is independent of its hemostatic effects. The finding that many viruses harness the host plasminogen to aid infectivity has suggested that antifibrinolytic agents may have antiviral benefits. Here, we review the broadening role of the plasminogen-activating system in physiology and pathophysiology and how manipulation of this system may be harnessed for benefits unrelated to its conventional application in thrombosis and hemostasis.


Asunto(s)
Plasminógeno/fisiología , Animales , Antifibrinolíticos/uso terapéutico , Encéfalo/enzimología , Conjuntivitis/fisiopatología , Activación Enzimática , Fibrina/metabolismo , Fibrinolisina/fisiología , Fibrinólisis/fisiología , Fibrinolíticos/uso terapéutico , Humanos , Inmunidad/fisiología , Infecciones/fisiopatología , Inflamación , Ratones , Plasminógeno/química , Plasminógeno/deficiencia , Plasminógeno/farmacología , Plasminógeno/uso terapéutico , Radiodermatitis/tratamiento farmacológico , Receptores de Superficie Celular/fisiología , Enfermedades Cutáneas Genéticas/fisiopatología , Trombosis/diagnóstico , Trombosis/tratamiento farmacológico , Ácido Tranexámico/farmacología , Ácido Tranexámico/uso terapéutico , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/fisiología , Heridas y Lesiones/tratamiento farmacológico
19.
J Biol Chem ; 296: 100099, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33208461

RESUMEN

Virulent strains of Streptococcus pyogenes (gram-positive group A Streptococcus pyogenes [GAS]) recruit host single-chain human plasminogen (hPg) to the cell surface-where in the case of Pattern D strains of GAS, hPg binds directly to the cells through a surface receptor, plasminogen-binding group A streptococcal M-protein (PAM). The coinherited Pattern D GAS-secreted streptokinase (SK2b) then accelerates cleavage of hPg at the R561-V562 peptide bond, resulting in the disulfide-linked two-chain protease, human plasmin (hPm). hPm localizes on the bacterial surface, assisting bacterial dissemination via proteolysis of host defense proteins. Studies using isolated domains from PAM and hPg revealed that the A-domain of PAM binds to the hPg kringle-2 module (K2hPg), but how this relates to the function of the full-length proteins is unclear. Herein, we use intact proteins to show that the lysine-binding site of K2hPg is a major determinant of the activation-resistant T-conformation of hPg. The binding of PAM to the lysine-binding site of K2hPg relaxes the conformation of hPg, leading to a greatly enhanced activation rate of hPg by SK2b. Domain swapping between hPg and mouse Pg emphasizes the importance of the Pg latent heavy chain (residues 1-561) in PAM binding and shows that while SK2b binds to both hPg and mouse Pg, the activation properties of streptokinase are strictly attributed to the serine protease domain (residues 562-791) of hPg. Overall, these data show that native hPg is locked in an activation-resistant conformation that is relaxed upon its direct binding to PAM, allowing hPm to form and provide GAS cells with a proteolytic surface.


Asunto(s)
Proteínas Bacterianas/metabolismo , Plasminógeno/química , Plasminógeno/metabolismo , Estreptoquinasa/química , Estreptoquinasa/metabolismo , Animales , Proteínas Bacterianas/química , Sitios de Unión , Humanos , Ratones , Unión Proteica , Infecciones Estreptocócicas/metabolismo , Virulencia
20.
Protein Sci ; 29(12): 2446-2458, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33058314

RESUMEN

Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) is an evolutionarily conserved essential enzyme in the glycolytic pathway. GAPDH is also involved in a wide spectrum of non-catalytic cellular 'moonlighting' functions. Bacterial surface-associated GAPDHs engage in many host interactions that aid in colonization, pathogenesis, and virulence. We have structurally and functionally characterized the recombinant GAPDH of the obligate intracellular bacteria Chlamydia trachomatis, the leading cause of sexually transmitted bacterial and ocular infections. Contrary to earlier speculations, recent data confirm the presence of glucose-catabolizing enzymes including GAPDH in both stages of the biphasic life cycle of the bacterium. The high-resolution crystal structure described here provides a close-up view of the enzyme's active site and surface topology and reveals two chemically modified cysteine residues. Moreover, we show for the first time that purified C. trachomatis GAPDH binds to human plasminogen and plasmin. Based on the versatility of GAPDH's functions, data presented here emphasize the need for investigating the Chlamydiae GAPDH's involvement in biological functions beyond energy metabolism.


Asunto(s)
Proteínas Bacterianas/química , Chlamydia trachomatis/enzimología , Gliceraldehído-3-Fosfato Deshidrogenasas/química , Modelos Moleculares , Plasminógeno/química , Proteínas Bacterianas/metabolismo , Cristalografía por Rayos X , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Plasminógeno/metabolismo , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA