Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(12)2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38928208

RESUMEN

Unfractionated heparin (UFH) and its low-molecular-weight fragments (LMWH) are widely used as anticoagulants for surgical procedures and extracorporeal blood purification therapies such as cardiovascular surgery and dialysis. The anticoagulant effect of heparin is essential for the optimal execution of extracorporeal blood circulation. However, at the end of these procedures, to avoid the risk of bleeding, it is necessary to neutralize it. Currently, the only antidote for heparin neutralization is protamine sulphate, a highly basic protein which constitutes a further source of serious side events and is ineffective in neutralizing LMWH. Furthermore, dialysis patients, due to the routine administration of heparin, often experience serious adverse effects, among which HIT (heparin-induced thrombocytopenia) is one of the most severe. For this reason, the finding of new heparin antagonists or alternative methods for heparin removal from blood is of great interest. Here, we describe the synthesis and characterization of a set of biocompatible macroporous cryogels based on poly(2-hydroxyethyl methacrylate) (pHEMA) and L-lysine with strong filtering capability and remarkable neutralization performance with regard to UFH and LMWH. These properties could enable the design and creation of a filtering device to rapidly reverse heparin, protecting patients from the harmful consequences of the anticoagulant.


Asunto(s)
Anticoagulantes , Criogeles , Heparina , Lisina , Heparina/química , Heparina/efectos adversos , Humanos , Criogeles/química , Anticoagulantes/química , Lisina/química , Heparina de Bajo-Peso-Molecular/química , Antagonistas de Heparina/química
2.
Biomacromolecules ; 18(12): 4113-4120, 2017 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-28949131

RESUMEN

The anticoagulant activity of heparin administered during medical interventions must be reversed to restore normal clotting, typically by titrating with protamine. Given the acute toxicity associated with protamine, we endeavored to generate safer heparin antagonists by engineering bacteriophage Qß virus-like particles (VLPs) to display motifs that bind heparin. A particle bearing a single amino acid change from wild-type (T18R) was identified as a promising candidate for heparin antagonism. Surface potential maps generated through molecular modeling reveal that the T18R mutation adds synergistically to adjacent positive charges on the particle surface, resulting in a large solvent-accessible cationic region that is replicated 180 times over the capsid. Chromatography using a heparin-sepharose column confirmed a strong interaction between heparin and the T18R particle. Binding studies using fluorescein-labeled heparin (HepFL) resulted in a concentration-dependent change in fluorescence intensity, which could be perturbed by the addition of unlabeled heparin. Analysis of the fluorescence data yielded a dissociation constant of approximately 1 nM and a 1:1 binding stoichiometry for HepFL:VLP. Dynamic light scattering (DLS) experiments suggested that T18R forms discrete complexes with heparin when the VLP:heparin molar ratios are equivalent, and in vitro clotting assays confirmed the 1:1 binding stoichiometry as full antagonism of heparin is achieved. Biolayer interferometry and backscattering interferometry corroborated the strong interaction of T18R with heparin, yielding Kd ∼ 1-10 nM. These biophysical measurements further validated T18R, and VLPs in general, for potential clinical use as effective, nontoxic heparin antagonists.


Asunto(s)
Allolevivirus/química , Antagonistas de Heparina/química , Heparina/química , Nanopartículas/química , Anticoagulantes/química , Sitios de Unión , Cápside/química , Proteínas de la Cápside/química , Cationes/química , Fluorescencia , Protaminas/química , Unión Proteica
3.
Prikl Biokhim Mikrobiol ; 52(4): 421-8, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-29512988

RESUMEN

Alkylated derivatives of low molecular weight chitosan with different substitution degrees of 98, 40, and 9% (I, II, and III respectively) have been synthesized. The structure of the obtained derivatives was defined by spectral assays (IR-spectroscopy and proton magnetic resonance). Chitosan derivatives were characterized with positive zeta-potential (33­51 mV) and solubility from 2 to 100 mg/mL in pH 7.4 and 25°C. It was shown that, at a concentration of 0.0014­0.0029 mg/mL, derivative I, as well as protamine sulfate, could be used to neutralize the anticoagulant activity of unfractionated or low molecular weight heparin. At a concentration of 0.0029­0.58 mg/mL, derivative I enhanced platelet aggregation, which would be necessary when hemostatic compounds or materials were used. Derivatives II and III enhanced platelet aggregation to a lesser extent.


Asunto(s)
Anticoagulantes , Plaquetas/metabolismo , Quitosano , Antagonistas de Heparina , Heparina , Agregación Plaquetaria/efectos de los fármacos , Anticoagulantes/química , Anticoagulantes/farmacología , Quitosano/análogos & derivados , Quitosano/química , Quitosano/farmacología , Heparina/química , Heparina/farmacología , Antagonistas de Heparina/química , Antagonistas de Heparina/farmacología , Humanos
4.
Adv Healthc Mater ; 13(20): e2400108, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38537246

RESUMEN

Thrombosis, the formation of blood clots within a blood vessel, can lead to severe complications including pulmonary embolism, cardiac arrest, and stroke. The most widely administered class of anticoagulants is heparin-based anticoagulants such as unfractionated heparin, low-molecular weight heparins (LMWHs), and fondaparinux. Protamine is the only FDA-approved heparin antidote. Protamine has limited efficacy neutralizing LMWHs and no reversal activity against fondaparinux. The use of protamine can lead to complications, including excessive bleeding, hypotension, and hypersensitivity, and has narrow therapeutic window. In this work, a new concept in the design of a universal heparin antidote: switchable protonation of cationic ligands, is presented. A library of macromolecular polyanion inhibitors (MPIs) is synthesized and screened to identify molecules that can neutralize all heparins with high selectivity and reduced toxicity. MPIs are developed by assembling cationic binding groups possessing switchable protonation states onto a polymer scaffold. By strategically selecting the identity and modulating the density of cationic binding groups on the polymer scaffold, a superior universal heparin reversal agent is developed with improved heparin-binding activity and increased hemocompatibility profiles leading to minimal effect on hemostasis. The activity of this heparin antidote is demonstrated using in vitro and in vivo studies.


Asunto(s)
Cationes , Heparina , Animales , Heparina/química , Heparina/farmacología , Ligandos , Cationes/química , Antagonistas de Heparina/química , Antagonistas de Heparina/farmacología , Humanos , Polielectrolitos/química , Polímeros/química , Antídotos/química , Antídotos/farmacología , Anticoagulantes/química , Anticoagulantes/farmacología , Ratones , Protaminas/química , Protaminas/farmacología
5.
Glycobiology ; 22(9): 1183-92, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22641771

RESUMEN

Slit3 is a large molecule with multiple domains and belongs to axon guidance families. To date, the biological functions of Slit3 are still largely unknown. Our recent study demonstrated that the N-terminal fragment of Slit3 is a novel angiogenic factor. In this study, we examined the biological function of the C-terminal fragment of human Slit3 (HSCF). The HSCF showed a high-affinity binding to heparin. The binding appeared to be heparin/heparan sulfate-specific and depends on the size, the degree of sulfation, the presence of N- and 6-O-sulfates and carboxyl moiety of the polysaccharide. Functional studies observed that HSCF inhibited antithrombin binding to heparin and neutralized the antifactor IIa and Xa activities of heparin and the antifactor IIa activity of low-molecular-weight heparin (LMWH). Thromboelastography analysis observed that HSCF reversed heparin's anticoagulation in global plasma coagulation. Taken together, these observations demonstrate that HSCF is a novel heparin-binding protein that potently neutralizes heparin's anticoagulation activity. This study reveals a potential for HSCF to be developed as a new antidote to treat overdosing of both heparin and LMWH in clinical applications.


Asunto(s)
Anticoagulantes/química , Antagonistas de Heparina/farmacología , Heparina/química , Heparitina Sulfato/química , Proteínas de la Membrana/química , Secuencia de Aminoácidos , Anticoagulantes/antagonistas & inhibidores , Antitrombina III/antagonistas & inhibidores , Antitrombina III/química , Sitios de Unión , Coagulación Sanguínea , Factor Xa/química , Inhibidores del Factor Xa , Antagonistas de Heparina/química , Heparina de Bajo-Peso-Molecular/antagonistas & inhibidores , Heparina de Bajo-Peso-Molecular/química , Heparitina Sulfato/antagonistas & inhibidores , Humanos , Proteínas de la Membrana/genética , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Unión Proteica , Estructura Terciaria de Proteína , Protrombina/antagonistas & inhibidores , Protrombina/química , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Soluciones , Tromboelastografía
6.
Chem Pharm Bull (Tokyo) ; 60(3): 371-6, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22382418

RESUMEN

Safer heparin-neutralizing agents are currently required to replace protamine, the use of which causes adverse effects such as anaphylaxia. Low-molecular-weight (LMW) heparin mimetics that potentiate antithrombin III (AT) action are also valuable as anti-thrombotics. This paper describes a high-throughput assay for both heparin-neutralizing agents and LMW heparin mimetics without the use of blood preparations. The assay is based on turbidimetric measurement of a solution of collagen, heparin, and a test compound. Native collagen molecules spontaneously form insoluble fibrils when transferred to a physiological buffer, and this process is inhibited by heparin. In the presence of a heparin-neutralizing agent or an LMW heparin mimetic, the inhibitory effect of heparin is canceled and turbidity increase is retrieved. We demonstrated that this assay is effective in detecting potential agents with high reliability (Z' factor=0.9). The screening of a chemical library (34400 compounds) was further performed in a 384-well format, and led to the identification of a novel heparin-neutralizing agent. Since this assay protocol is feasible for an automated high-throughput screening (HTS) system, it could enhance the lead seeking process for drugs related to heparin/heparan sulfate (HS) functions.


Asunto(s)
Antagonistas de Heparina/química , Heparina de Bajo-Peso-Molecular/química , Antitrombina III/antagonistas & inhibidores , Colágeno/química , Antagonistas de Heparina/farmacología , Heparina de Bajo-Peso-Molecular/farmacología , Heparitina Sulfato/química , Peso Molecular , Nefelometría y Turbidimetría/métodos , Protaminas/efectos adversos , Reproducibilidad de los Resultados , Soluciones/química
7.
J Vasc Surg ; 54(3): 791-8, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21620612

RESUMEN

OBJECTIVES: The localized delivery of exogenous, angiogenic growth factors such as fibroblast growth factor (FGF)-2 has become a promising alternative treatment of peripheral artery disease (PAD) and critical limb ischemia (CLI). The present study describes the efficacy of fragmin/protamine microparticles containing FGF-2 (F/P-MPs/FGF-2) to promote vessel growth in a rabbit model of hindlimb ischemia. METHODS: A total of 24 rabbits were used to construct a model of hindlimb ischemia by resection of the left femoral artery. The rabbits were randomly divided into four groups 10 days after surgery (day 0); group A: control (non-treated; 1 mL of phosphate-buffered saline [PBS]); group B: FGF-2 (100 µg FGF-2 in 1 mL PBS)-treated; group C: F/P-MPs (12 mg dried F/P MPs in 1 mL PBS)-treated; and group D; F/P MPs/FGF-2 (100 µg FGF-2 and 12 mg dried F/P MPs in 1 mL PBS)-treated (n = 6 each). The drugs were administered intramuscularly to each group. Blood flow and blood pressure were measured in each group on days 0, 14, and 28. Angiography was performed to assess arteriogenesis on day 28. The number of capillaries on day 28 was determined by direct counting CD31(-) and α-smooth muscle antibody (α-SMA)-positive vessels. RESULTS: Neither death nor wound infection was observed throughout the experiment. The F/P MPs/FGF-2-treated group showed marked improvement in the blood flow ratio, blood pressure ratio, and capillary number in comparison to the control group, FGF-2-treated group, and F/P MPs-treated group. The F/P MPs-treated group showed intermediate improvement in blood flow ratio and capillary number in comparison to the control group and FGF-2-treated group. CONCLUSIONS: The F/P MPs/FGF-2-treated group strongly induced functional collateral vessels in the rabbit model of hindlimb ischemia, indicating a possible therapy for PAD.


Asunto(s)
Inductores de la Angiogénesis/administración & dosificación , Anticoagulantes/química , Circulación Colateral/efectos de los fármacos , Dalteparina/química , Factor 2 de Crecimiento de Fibroblastos/administración & dosificación , Antagonistas de Heparina/química , Isquemia/tratamiento farmacológico , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica/efectos de los fármacos , Protaminas/química , Actinas/metabolismo , Análisis de Varianza , Inductores de la Angiogénesis/química , Animales , Presión Sanguínea/efectos de los fármacos , Capilares/efectos de los fármacos , Capilares/metabolismo , Capilares/fisiopatología , Química Farmacéutica , Modelos Animales de Enfermedad , Portadores de Fármacos , Composición de Medicamentos , Factor 2 de Crecimiento de Fibroblastos/química , Miembro Posterior , Inmunohistoquímica , Inyecciones Intramusculares , Isquemia/diagnóstico por imagen , Isquemia/metabolismo , Isquemia/fisiopatología , Flujometría por Láser-Doppler , Masculino , Tamaño de la Partícula , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Conejos , Radiografía , Flujo Sanguíneo Regional/efectos de los fármacos , Factores de Tiempo
8.
Bioorg Med Chem ; 19(3): 1131-5, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20719523

RESUMEN

A new approach to enzyme-responsive MRI agents based on the use of liposomes loaded with a high number of paramagnetic metal complexes (Gd-HPDO3A) is presented. It relies on the disruption of low relaxivity aggregates formed by liposomes and a macromolecular substrate that is selectively cleaved by the enzyme of interest. The interaction of anionic liposomes composed of POPC:CHOL:DPGS and the cationic protein protamine yields a poorly soluble supramolecular assembly endowed with a low relaxivity. The action of the serine protease trypsin causes the digestion of protamine and the consequent de-assembly of the supramolecular adduct. The process is accompanied by an overall relaxation enhancement of solvent water protons as consequence of the dissolution of the aggregated liposomes. The observed increase of relaxivity is linearly dependent on the enzyme concentration. An illustrative example of the possible use of the herein presented responsive agent has been reported. It consists of the entrapment of the supramolecular assembly in alginate microcapsules that have often been used as envelopes for in vivo applications of stem cells and pancreatic islets. The change in the observed longitudinal relaxation rate R(1) (leading to an hyperintense signal in the corresponding MR images) may act as a sensor of the protease activity in the biological environment in which the capsules is located.


Asunto(s)
Compuestos Heterocíclicos/síntesis química , Liposomas/química , Sondas Moleculares/química , Compuestos Organometálicos/síntesis química , Péptido Hidrolasas/química , Medios de Contraste/química , Gadolinio/química , Antagonistas de Heparina/química , Compuestos Heterocíclicos/química , Imagen por Resonancia Magnética/métodos , Compuestos Organometálicos/química , Péptido Hidrolasas/metabolismo , Permeabilidad , Protaminas/química , Protones , Tripsina/química , Agua/química
9.
J Nanosci Nanotechnol ; 11(4): 3058-64, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21776671

RESUMEN

We synthesized a nanoparticle (NP) for ex-vivo cell labeling and MRI tracking by covalently coupling the C-terminus of a rhodamine-labeled protamine (ProRho) to Feraheme (FH) in order to yield the nanoparticle denoted ProRho-FH. Since protamine can adsorb to certain charged surfaces, we confirmed a covalent interaction between ProRho and FH by heparin affinity chromatography. ProRho-FH lacks a net charge (zeta potential approximately 0) due to the combination of negative FH and positive ProRho charges. ProRho-FH was readily internalized by U87 cells and mouse mesenchymal stem cells as determined by FACS and MR relaxometry. Finally, some 4,000 stem cells were implanted in a mouse brain and imaged by MRI. Due to its lack of net surface charge, ProRho-FH relies on the internalizing properties of the surface guanidinium groups present in the arginine-rich protamine to induce NP uptake. ProRho-FH is a unique cell-labeling agent due to its synthesis using two approved drugs, magnetofluorescence, site-specific covalent attachment chemistry, and lack of surface charge.


Asunto(s)
Rastreo Celular/métodos , Imagen por Resonancia Magnética/métodos , Nanopartículas de Magnetita , Nanocápsulas/química , Protaminas/administración & dosificación , Células Madre/citología , Animales , Medios de Contraste , Hematínicos/administración & dosificación , Hematínicos/química , Antagonistas de Heparina/administración & dosificación , Antagonistas de Heparina/química , Ratones , Microscopía Fluorescente/métodos , Protaminas/química , Trasplante de Células Madre
10.
FEBS Open Bio ; 11(9): 2468-2489, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34184429

RESUMEN

Unfractionated heparin (UFH) is an anionic glycosaminoglycan that is widely used to prevent blood clotting. However, in certain cases, unwanted side effects can require it to be neutralized. Protamine sulfate (PS), a basic peptide rich in arginine, is the only approved antagonist for UFH neutralization. Many adverse reactions occur with the clinical application of PS, including systemic hypotension, pulmonary hypertension, and anaphylaxis. We previously described R15, a linear peptide composed of 15 arginine molecules, as a potential UFH antagonist. In this study, the in-depth safety of R15 was explored to reveal its merits and associated risks in comparison with PS. In vitro safety studies investigated the interactions of R15 with erythrocytes, fibrin, complement, and rat plasma. In vivo safety studies explored potential toxicity and immunogenicity of R15 and the UFH-R15 complex. Results showed that both PS and R15 can induce erythrocyte aggregation, thicken fibrin fibers, activate complement, and cause anticoagulation in a concentration-dependent manner. However, those influences weakened in whole blood or in live animals and were avoided when R15 was in a complex with UFH. We found dramatically enhanced complement activation when there was excess UFH in analyses involving UFH-PS complexes, and a slight increase in those involving UFH-R15 complexes. Within 2 h, R15 was degraded in rat plasma in vitro, whereas PS was not. Enhanced creatinine was found after a single intravenous injection of PS or R15 (900 U·kg-1 , body weight), suggesting possible abnormal renal function. The UFH-PS complex, but not the UFH-R15 complex, exhibited obvious immunogenicity. In conclusion, R15 is nonimmunogenic and potentially safe at a therapeutic dose to reverse the effects of UFH.


Asunto(s)
Antagonistas de Heparina/farmacología , Heparina/farmacología , Péptidos/farmacología , Animales , Coagulación Sanguínea/efectos de los fármacos , Pruebas de Coagulación Sanguínea , Activación de Complemento , Antagonismo de Drogas , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Fibrina/metabolismo , Antagonistas de Heparina/química , Humanos , Péptidos/química , Protaminas/química , Protaminas/farmacología , Ratas
11.
Transfus Med Rev ; 21(2): 164-9, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17489141

RESUMEN

Doctor Parviz Lalezari, currently a clinical professor of Medicine and Pathology at Albert Einstein College of Medicine in New York, describes highlights of his research career since 1958. He became the director of the blood bank at Montefiore Hospital in New York City in 1961, director of the Division of Immunohematology until 1996, and then until 2001, was President and chief executive officer of the Bergen Community Regional Blood Center in New Jersey. Doctor Lalezari was born in Iran in 1931, and after graduation from Medical School, he came to the United States in 1956. His initial research was on leukocyte antibodies. After modifying the available antibody detection techniques, he discovered that like hemolytic disease of the newborn and neonatal immune thrombocytopenia, fetal-maternal neutrophil incompatibility can cause neonatal neutropenia. He identified the targets of these antibodies and showed that they were expressed only on peripheral blood neutrophils. Doctor Lalezari also discovered that a common form of neutropenia in early childhood was caused by development of autoantibodies, which surprisingly were directed against the same neutrophil-specific antigens involved in fetal-maternal incompatibility. In 1959, a heparin-neutralizing drug (Polybrene) was introduced to be used after open-heart surgery. Lalezari discovered that Polybrene, a quaternary ammonium polymer, reacted with sialic acid molecules on the red blood cell (RBC) surface, causing the RBCs to aggregate. Later, realizing that the repelling forces generated by the RBC surface membrane charges were responsible for failure of the small IgG antibody molecules to agglutinate the RBCs, he used Polybrene to neutralize the RBC surface negative charge to allow the IgG antibody molecules to induce hemagglutination. This became The Polybrene test, which is to be used in RBC antibody detection.


Asunto(s)
Autoanticuerpos , Agregación Eritrocitaria , Leucocitos , Púrpura Trombocitopénica Idiopática , Sangrado por Deficiencia de Vitamina K , Autoanticuerpos/historia , Autoanticuerpos/inmunología , Agregación Eritrocitaria/inmunología , Antagonistas de Heparina/química , Antagonistas de Heparina/historia , Bromuro de Hexadimetrina/química , Bromuro de Hexadimetrina/historia , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Recién Nacido , Leucocitos/inmunología , Púrpura Trombocitopénica Idiopática/historia , Púrpura Trombocitopénica Idiopática/inmunología , Sangrado por Deficiencia de Vitamina K/historia , Sangrado por Deficiencia de Vitamina K/inmunología
12.
Int J Pharm ; 306(1-2): 132-41, 2005 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-16246511

RESUMEN

Heparin employed in extracorporeal blood circulation (ECBC) procedures (e.g. open heart operations) often leads to a high incidence of bleeding complications. Protamine employed in heparin neutralization, on the other hand, can cause severe adverse reactions. We previously developed an approach that could prevent both heparin- and protamine-induced toxic side effects concomitantly. This approach consisted of placing a hollow fiber-based bioreactor device containing immobilized protamine (termed a "protamine bioreactor") at the distal end of the ECBC procedure. This protamine bioreactor would remove heparin after heparin served its anticoagulant purpose in the ECBC device, thereby eliminating heparin-induced bleeding risks. In addition, this protamine bioreactor would prevent protamine from entering the patients, thereby aborting any protamine-induced toxic effects. Both in vitro and in vivo studies have successfully demonstrated the feasibility of this approach. Despite promises, early findings also revealed two shortcomings that must be overcome for the protamine bioreactor to be applied clinically. The first drawback was that the cyanate ester linkages, involved in conjugating protamine to the bioreactor device, were unstable and prone to hydrolysis, resulting in the leakage of a significant amount of protamine into circulation during application of the protamine bioreactor. The second deficiency was that the capacity of the protamine bioreactor in heparin removal was rather low, owing to the limited surface area of the hollow fibers for protamine immobilization and subsequently heparin adsorption. In this paper, we present novel strategies to overcome these two limitations. A new conjugation method based on the use of 4-(oxyacetyl)phenoxyacetic acid (OAPA) as the activating reagent was employed to yield stable linkages, via the abundant arginine residues of protamine, onto the hollow fibers. Results showed that while the amount of protamine immobilized on each gram of fibers was relatively comparable between the OAPA and the previous CNBr activation methods (7.45 mg/g versus 7.69 mg/g fibers), there was virtually no detectable leaching of immobilized protamine from the bioreactor by the OAPA method, comparing to 35% leaching of protamine by the previous CNBr method following 72 h of storage of the bioreactor in PBS buffer at 37 degrees C. To improve the capacity and functionality of the protamine bioreactor, two novel approaches were adopted. Long chain and high molecular weight poly-lysine was linked to the hollow fibers, prior to protamine coupling, to create multiple layers of immobilized protamine for subsequent heparin adsorption. In addition, a poly(ethylene glycol) (PEG) chain was inserted between protamine and the hollow fibers to yield a three-dimensional, free dynamic motion for immobilized protamine. Preliminary observations indicated that a four- to five-fold enhancement in heparin adsorption was attained by utilizing each of these new approaches. Aside from their current use, these new strategies can also be employed generically to improve the functionality of any affinity-type bioreactor. Indeed, efforts have been made recently in utilizing these approaches to develop a clinically usable GPIIb/IIIa bioreactor for the treatment of immune thrombocytopenic purpura (ITP)-an autoimmune disease.


Asunto(s)
Anticoagulantes/química , Circulación Extracorporea/instrumentación , Glicolatos/química , Antagonistas de Heparina/química , Heparina/química , Protaminas/química , Anticoagulantes/efectos adversos , Anticoagulantes/farmacología , Diseño de Equipo , Heparina/efectos adversos , Heparina/farmacología , Antagonistas de Heparina/efectos adversos , Antagonistas de Heparina/farmacología , Estructura Molecular , Polietilenglicoles/química , Polilisina/química , Protaminas/efectos adversos , Protaminas/farmacología
13.
Thromb Res ; 135(1): 127-9, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25466847

RESUMEN

INTRODUCTION: Since heparin-induced thrombocytopenia (HIT), caused by the generation of antibodies against platelet factor 4 (PF4)/heparin complexes (HIT antibodies), may induce serious complications due to thrombosis, a prompt diagnosis is desirable. Functional tests with platelet activation to detect HIT antibodies are useful for diagnosis of HIT, in particular (14)C-selotonin release assay (SRA). However, they are complicated and so can be performed only in limited laboratories. We tested if a blood coagulation test using Sonoclot® analyzer can serve for the detection of HIT antibodies. MATERIALS AND METHODS: A murine monoclonal antibody (HIT-MoAb) against PF4/heparin complexes was used as an alternative to human HIT antibodies. To the mixture of HIT-MoAb and heparin (0.5 U/mL, final), whole blood obtained from a healthy volunteer was added, and then the activated clotting time (ACT), clot rate (CR), and area under the curve (AUC) were measured with Sonoclot® analyzer for 30minutes. RESULTS: The HIT-MoAb (30 to 100µg/mL, final) concentration dependently suppressed the anticoagulation activity (prolongation of ACT and decrease of CR and AUC) of heparin. CONCLUSIONS: The suppression of anticoagulation effect of heparin by HIT-MoAb was demonstrated by measurements using Sonoclot® analyzer. This method may provide a new tool for screening of HIT antibodies.


Asunto(s)
Anticuerpos/química , Coagulación Sanguínea , Antagonistas de Heparina/química , Trombocitopenia/inmunología , Animales , Anticuerpos Monoclonales de Origen Murino/química , Anticoagulantes/química , Área Bajo la Curva , Pruebas de Coagulación Sanguínea , Relación Dosis-Respuesta a Droga , Voluntarios Sanos , Heparina/química , Humanos , Ratones , Activación Plaquetaria , Factor Plaquetario 4/inmunología , Trombocitopenia/inducido químicamente , Trombosis/inmunología
14.
Surgery ; 121(4): 419-24, 1997 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9122872

RESUMEN

BACKGROUND: Recent studies have shown that patients with heparin-induced thrombocytopenia (HIT) form immunoglobulin G (IgG) and/or IgM antibodies directed against a complex of platelet factor 4 (PF4) and heparin. This recognition has resulted in the development of enzyme-linked immunosorbent assays (ELISAs) that use the heparin/PF4 complex as the antigen. This study describes the use of a standardized ELISA to assess antibody formation in five patients suspected of having HIT. METHODS: Five patients received heparin for treatment of arterial or venous thrombotic disorders. All patients had the ELISA performed to detect IgG or IgM antibodies directed against heparin-PF4, as well as the 14C serotonin release assay, when HIT was clinically suspected. RESULTS: HIT was diagnosed in four patients and ruled out in a fifth by using the ELISA. All patients had a 40% decrease in platelet count that returned to normal after heparin cessation. Only one of the four patients who tested positive by ELISA for IgG antibodies also tested positive by the 14C serotonin release assay. Treatment was significantly altered by the ELISA results in all five patients. CONCLUSIONS: It is likely that the ELISA is more sensitive in the diagnosis of HIT than the more traditional aggregation tests, and it may emerge as a new gold standard. Prospective studies in which serial laboratory testing is combined with measurement of clinical outcomes are needed and will eventually provide a greater understanding of the full spectrum of HIT and the clinical settings that precipitate thrombosis in the vascular surgery patient.


Asunto(s)
Fibrinolíticos/efectos adversos , Heparina/efectos adversos , Trombocitopenia/inducido químicamente , Procedimientos Quirúrgicos Vasculares , Adulto , Ensayo de Inmunoadsorción Enzimática , Femenino , Heparina/química , Antagonistas de Heparina/química , Humanos , Masculino , Persona de Mediana Edad , Recuento de Plaquetas , Factor Plaquetario 4/química , Trombocitopenia/diagnóstico
15.
Thromb Res ; 111(4-5): 273-9, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14693175

RESUMEN

A lack of correlation between activated partial thromboplastin time (aPTT), thrombin time (TT) and anti-factor Xa (AXa) activity was observed in patients after cardiac surgery with cardiopulmonary bypass (CBP). Indeed, AXa activity measured by the chromogenic assay, Coamatic Heparin, was higher than expected with regard to results obtained in coagulation assays. To account for this discrepancy, another AXa chromogenic assay was tested. First, AXa activity was measured with two chromogenic assays (Coamatic Heparin and Rotachrom Heparin) in plasma samples of 25 patients undergoing cardiac surgery at two time points after heparin reversal by protamine. AXa activity was significantly higher when measured with Coamatic Heparin than with Rotachrom Heparin in samples collected just after protamine infusion (p<0.01). Next, since Coamatic( Heparin contains dextran sulfate (DXS) to reduce the influence of heparin antagonists such as platelet factor 4 (PF4), whereas Rotachrom Heparin does not, we hypothesized that the dextran sulfate contained in the reagent might explain this discrepancy. We therefore performed in vitro studies consisting in neutralizing unfractionated heparin (UFH) with protamine and measuring AXa activity with the two chromogenic assays. An AXa activity was still measurable with Coamatic Heparin after neutralization, thus strongly suggesting that dextran sulfate dissociates protamine/heparin complexes. We conclude that Coamatic Heparin assays should be avoided when measuring AXa activity in plasma samples immediately after protamine infusion, as inaccurate results may lead to inadequate management of heparin reversal.


Asunto(s)
Sulfato de Dextran/sangre , Factor Xa/análisis , Heparina/sangre , Protaminas/sangre , Anticoagulantes/sangre , Anticoagulantes/química , Anticoagulantes/uso terapéutico , Pruebas de Coagulación Sanguínea/métodos , Procedimientos Quirúrgicos Cardiovasculares/efectos adversos , Compuestos Cromogénicos/análisis , Compuestos Cromogénicos/química , Sulfato de Dextran/química , Factor Xa/química , Heparina/química , Heparina/uso terapéutico , Antagonistas de Heparina/sangre , Antagonistas de Heparina/química , Antagonistas de Heparina/uso terapéutico , Humanos , Indicadores y Reactivos/química , Protaminas/química , Protaminas/uso terapéutico , Juego de Reactivos para Diagnóstico , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Trombosis/etiología , Trombosis/prevención & control
16.
Life Sci ; 73(22): 2793-806, 2003 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-14511765

RESUMEN

In the blood coagulation cascade, human antithrombin III (hAT III) acts as an inhibitor of serine proteases such as thrombin and factor Xa, and this anticoagulatory glycoprotein requires the binding of heparin for its activation. In this study, we synthesized the polypeptides corresponding to the proposed heparin-binding sites including the (41-49), (286-301) and (123-139) regions of hAT III, and examined their interactions with heparin by means of physicochemical and biochemical methods. All the synthetic peptides had a high affinity toward heparin, evidenced by the fact that they were eluted from a heparin-agarose column at the high salt concentration range of 520-700 mM. In addition, hAT III (123-139) attenuated the effect of heparin on the activation of hAT III, whereas other HBPs did not, suggesting that only hAT III (123-139) could interact with the active site of heparin. On the basis of these results, we prepared novel hAT III (123-139)-related derivatives as potent heparin antagonist candidates, and examined the influence of several modifications on their activity in vitro. The results provided new findings about the structure-activity relationship of hAT III (123-139), and led us to the successful development of a potent antagonist for heparin.


Asunto(s)
Anticoagulantes/antagonistas & inhibidores , Antitrombina III/análogos & derivados , Antitrombina III/farmacología , Antagonistas de Heparina/farmacología , Heparina/metabolismo , Péptidos/farmacología , Anticoagulantes/química , Anticoagulantes/metabolismo , Antitrombina III/química , Fenómenos Químicos , Química Física , Cromatografía en Agarosa , Dicroismo Circular , Dalteparina/metabolismo , Inhibidores del Factor Xa , Heparina/química , Antagonistas de Heparina/química , Humanos , Modelos Moleculares , Péptidos/química , Unión Proteica
17.
Int J Pharm ; 283(1-2): 11-7, 2004 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-15363497

RESUMEN

Protamine is a well-known excipient in pharmaceutics. It represents a peptide consisting of exclusive aliphatic amino acids, hence it cannot be quantified by UV-spectroscopy (lambdamax 280 nm). A new and sensitive quantification method based on the derivatisation of protamine with ortho-phthaldialdehyde (OPA) in the presents of 2-mercaptoethanol (ME) or N-acetyl-L-cysteine (NAC) in basic aqueous solution using 96-well microtiter plates are introduced in this report. The resulting isoindol derivatives reveal a fluorescence excitation (maximum lambdaex 345 nm) and emission (maximum lambdaem 450 nm) spectra. Derivatives of OPA/NAC reagent were found to be useful for protamine quantification in pharmaceutical nanoparticle preparation containing DNA. A sufficient stability of the isoindol derivatives was shown. It was possible to determine protamine free base, protamine sulphate and protamine chloride with limits of detection less than 1.1 microg/ml.


Asunto(s)
Antagonistas de Heparina/química , Protaminas/química , Tecnología Farmacéutica , Acetilcisteína , Indicadores y Reactivos , o-Ftalaldehído
18.
AAPS PharmSci ; 3(3): E17, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11741268

RESUMEN

Low molecular weight protamine (LMWP) appears to be a promising solution for heparin neutralization without the protamine-associated catastrophic toxic effects. The feasibility of this hypothesis was proven previously by using a peptide mixture produced from proteolytic digestion of protamine. To further examine the utility of this compound as an ultimate nontoxic protamine substitute, detailed studies on the purification and characterization of LMWP including the precise amino acid sequence, structure-function relationship, and possible mechanism were conducted. A number of LWMP fragments, composed of highly cationic peptides with molecular weights ranging from 700 to 1900 d, were prepared by digestion of native protamine with the protease thermolysin. These fragments were fractionated using a heparin affinity chromatography, and their relative binding strengths toward heparin were elucidated. Five distinct fractions were eluted at NaCl concentration ranging from 0.4 to 1.0 M and were denoted as TDSP1 to TDSP5, in increasing order of eluting ionic strength. Among these 5 fractions, TDSP4 and TDSP5 contained 3 LMWP peptide fragments, and they were found to retain the complete heparin-neutralizing function of protamine. By using a peptide mass spectrometry (MS) fingerprint mapping technique, the amino acid sequences of the microheterogeneous LMWP fragments in all these 5 elution fractions were readily identified. A typical structural scaffold made by arginine clusters in the middle and nonarginine residues at the N-terminal of the peptide sequence was observed for all these LMWP fragments. By aligning the sequences with the potency in heparin neutralization of these LMWP fragments, it was found that retention of potency similar to that of protamine required the presence of at least 2 arginine clusters in the LMWP fragments; such as the sequence of VSRRRRRRGGRRRR seen in the most potent LMWP fraction-TDSP5. The above finding was further validated by using a synthetic LMWP analogue-CRRRRRRR-and it was found that its heparin-neutralizing ability was increased by changing from a monomeric to a dimeric structure of this analogue peptide. Based on these results, the structural requirement for a compound to function as an effective heparin antidote and the possible mechanism involved in heparin neutralization were established.


Asunto(s)
Anticoagulantes/química , Antagonistas de Heparina/química , Heparina/química , Fragmentos de Péptidos/química , Protaminas/química , Anticoagulantes/metabolismo , Antitrombina III/metabolismo , Unión Competitiva , Cromatografía de Afinidad , Dimerización , Heparina/metabolismo , Antagonistas de Heparina/aislamiento & purificación , Antagonistas de Heparina/metabolismo , Humanos , Técnicas In Vitro , Peso Molecular , Fragmentos de Péptidos/aislamiento & purificación , Fragmentos de Péptidos/metabolismo , Análisis de Secuencia de Proteína , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Relación Estructura-Actividad , Termolisina
19.
AAPS PharmSci ; 3(3): E19, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11741270

RESUMEN

Heparin employed in cardiovascular surgeries often leads to a high incidence of bleeding complications. Protamine employed in heparin reversal, however, can cause severe adverse reactions. In an attempt to address this clinical problem, we developed low molecular weight protamine (LMWP) as a potentially effective and less toxic heparin antagonist. A homogeneous 1880-d peptide fragment, termed LMWP-TDSP5 and containing the amino acid sequence of VSRRRRRRGGRRRR, was derived directly from protamine by enzymatic digestion of protamine with thermolysin. In vitro studies demonstrated that TDSP5 was capable of neutralizing various anticoagulant functions of both heparin and commercial low molecular weight heparin preparations. In addition, TDSP5 exhibited significantly reduced crossreactivity toward mouse sera containing antiprotamine antibodies. TDSP5 showed a decrease in its potential in activating the complement system. All of these findings suggested the possibility of markedly reduced protamine toxicity for TDSP5. In this article, we conducted preliminary in vivo studies to further demonstrate the feasibility and utility of using LMWP as a nontoxic clinical protamine substitute. Dogs were chosen as test animals because they were known to magnify the typical human response to protamine. By using a full spectra of biological and clinical assays for heparin, including the anti-IIa and anti-Xa chromogenic assays and the activated partial, thromboplastin time and TCT clotting assays, TDSP5 showed that it could completely neutralize all these different anticoagulant functions of heparin in dogs. Although administration of protamine in dogs produced a significant reduction in mean arterial blood pressure (-14.9 mm Hg) and elevation in pulmonary artery systolic pressure (+5.0 mm Hg), the use of TDSP5 in dogs did not elicit any statistically significant change in any of the variables measured. Furthermore, the use of LMWP also significantly reduced the protamine-induced transient thrombocytopenic and granulocytopenic responses. The white blood cell counts and platelet counts decreased to 82.1% and 60.0% of baseline, respectively, in dogs given intravenous protamine compared to 97.8% and 88.6% of baseline in dogs receiving TDSP5. These preliminary findings indicated that LMWP could potentially provide an effective and safe means to control both heparin- and protamine-induced complications.


Asunto(s)
Anticoagulantes/toxicidad , Antagonistas de Heparina/farmacología , Heparina/toxicidad , Fragmentos de Péptidos/farmacología , Protaminas/química , Animales , Recuento de Células Sanguíneas , Coagulación Sanguínea/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Perros , Inhibidores del Factor Xa , Antagonistas de Heparina/química , Antagonistas de Heparina/toxicidad , Masculino , Fragmentos de Péptidos/química , Fragmentos de Péptidos/toxicidad , Protrombina/antagonistas & inhibidores , Tiempo de Protrombina , Presión Esfenoidal Pulmonar/efectos de los fármacos
20.
J Control Release ; 193: 63-73, 2014 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-24943246

RESUMEN

Low molecular weight protamine (LMWP) is a peptide fragment produced in our laboratory from enzymatic digestion of native protamine. More than 30 papers studying the properties and applications of LMWP have been published by our group in various journals since its initial discovery in 1999. Results have shown that LMWP could completely neutralize the anticoagulant functions of both heparin and low molecular weight heparin (LMWH), with reduced antigenicity and cross-reactivity toward the mice-derived anti-protamine antibodies. Aside from its potential as a heparin/LMWH antagonist, LMWP also shows the ability to retard insulin adsorption by the formation of an insoluble complex, making it a less toxic long-lasting insulin product than the conventional neutral protamine Hagedorn (NPH) insulin for diabetic control. Importantly, LMWP (Sequence: VSRRRRRRGGRRRR), with 10 arginine residues in its structure, could function as a cell-penetrating peptide (CPP), also termed protein transduction domain (PTD), to achieve effective intracellular protein or gene delivery in clinical practice. In this paper, we present a thorough review of our work related to LMWP, with the aim of providing readers an insight into its potential to be a clinical protamine substitute as well as a non-toxic cell penetrating peptide applicable to achieve intracellular protein and gene delivery.


Asunto(s)
Péptidos de Penetración Celular/química , Antagonistas de Heparina/química , Fragmentos de Péptidos/química , Protaminas/química , Animales , Anticuerpos/sangre , Péptidos de Penetración Celular/inmunología , Péptidos de Penetración Celular/farmacología , Péptidos de Penetración Celular/toxicidad , Reacciones Cruzadas/efectos de los fármacos , Reacciones Cruzadas/inmunología , Composición de Medicamentos , Antagonistas de Heparina/farmacología , Antagonistas de Heparina/toxicidad , Ratones , Peso Molecular , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/farmacología , Fragmentos de Péptidos/toxicidad , Protaminas/inmunología , Protaminas/farmacología , Protaminas/toxicidad , Conformación Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA