Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cells ; 13(10)2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38786070

RESUMEN

Sickle cell disease (SCD) is the most common genetic blood disorder in the United States, with over 100,000 people suffering from this debilitating disease. SCD is caused by abnormal hemoglobin (Hb) variants that interfere with normal red blood cell (RBC) function. Research on SCD has led to the development and approval of several new SCD therapies in recent years. The recent FDA-approved novel gene therapies are potentially curative, giving patients an additional option besides a hematopoietic bone marrow transplant. Despite the promise of existing therapies, questions remain regarding their long-term pharmacological effects on adults and children. These questions, along with the exorbitant cost of the new gene therapies, justify additional research into more effective therapeutic options. Continual research in this field focuses on not only developing cheaper, more effective cures/treatments but also investigating the physiological effects of the current therapies on SCD patients, particularly on the brain and kidneys. In this article, we undertake a comprehensive review of ongoing clinical trials with completion dates in 2024 or later. Our exploration provides insights into the landscape of current therapeutics and emerging novel therapies designed to combat and potentially eradicate SCD, including the latest FDA-approved gene therapies.


Asunto(s)
Anemia de Células Falciformes , Terapia Genética , Humanos , Anemia de Células Falciformes/terapia , Anemia de Células Falciformes/genética , Ensayos Clínicos como Asunto
2.
Nucleic Acid Ther ; 33(4): 277-286, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37093131

RESUMEN

The leading cause of death in adults in the United States is cardiovascular disease, with mortality and morbidity mainly attributed to thromboembolism. Heparin is the most common therapy used for treating venous and arterial thrombosis. Heparin effectively accelerates the inhibition of coagulation proteases thrombin and factor Xa through the serine protease inhibitor (serpin) antithrombin (AT). Heparin is an essential therapeutic anticoagulant because of its effectiveness and the availability of protamine sulfate as an antidote. However, heparin therapy has several limitations. Thus, new anticoagulants, including direct thrombin inhibitors (ie, argatroban) and low-molecular-weight heparins (ie, fondaparinux), are used to treat some thromboembolic disorders. We developed and characterized a family of novel RNA-based aptamers that bind AT using two novel selection schemes. One of the aptamers, AT-16, accelerates factor Xa inhibition by AT in the absence of heparin. AT-16's effect on thrombin inhibition by AT is less effective compared to factor Xa. AT-16 induces a conformational change in AT that is different from that induced by heparin. This study demonstrates that an AT-specific RNA aptamer, AT-16, exhibits a positive allosteric modulator effect on AT's inhibition of factor Xa.


Asunto(s)
Aptámeros de Nucleótidos , Aptámeros de Nucleótidos/genética , Aptámeros de Nucleótidos/farmacología , Factor Xa , Trombina , Anticoagulantes/farmacología , Heparina/uso terapéutico , Antitrombinas/farmacología
3.
Int J Mol Sci ; 22(8)2021 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-33918821

RESUMEN

Aptamers are single-stranded DNA or RNA sequences that bind target molecules with high specificity and affinity. Aptamers exhibit several notable advantages over protein-based therapeutics. Aptamers are non-immunogenic, easier to synthesize and modify, and can bind targets with greater affinity. Due to these benefits, aptamers are considered a promising therapeutic candidate to treat various conditions, including hematological disorders and cancer. An active area of research involves developing aptamers to target blood coagulation factors. These aptamers have the potential to treat cardiovascular diseases, blood disorders, and cancers. Although no aptamers targeting blood coagulation factors have been approved for clinical use, several aptamers have been evaluated in clinical trials and many more have demonstrated encouraging preclinical results. This review summarized our knowledge of the aptamers targeting proteins involved in coagulation, anticoagulation, fibrinolysis, their extensive applications as therapeutics and diagnostics tools, and the challenges they face for advancing to clinical use.


Asunto(s)
Aptámeros de Nucleótidos/farmacología , Factores de Coagulación Sanguínea/genética , Coagulación Sanguínea , Marcación de Gen , Animales , Aptámeros de Nucleótidos/química , Aptámeros de Nucleótidos/uso terapéutico , Coagulación Sanguínea/efectos de los fármacos , Coagulación Sanguínea/fisiología , Factores de Coagulación Sanguínea/metabolismo , Proteínas Portadoras , Evaluación Preclínica de Medicamentos , Fibrinólisis , Marcación de Gen/métodos , Humanos , Unión Proteica , Técnica SELEX de Producción de Aptámeros , Transducción de Señal
4.
Nucleic Acid Ther ; 27(6): 354-364, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29039727

RESUMEN

The pathophysiology of sickle cell disease (SCD) is dependent on the polymerization of deoxygenated sickle hemoglobin (HbS), leading to erythrocyte deformation (sickling) and vaso-occlusion within the microvasculature. Following deoxygenation, there is a delay time before polymerization is initiated, during which nucleation of HbS monomers occurs. An agent with the ability to extend this delay time or slow polymerization would therefore hold a therapeutic, possibly curative, potential. We used the Systematic Evolution of Ligands by Exponential Enrichment (SELEX) method to screen for HbS-binding RNA aptamers modified with nuclease-resistant 2'-fluoropyrimidines. Polymerization assays were employed to identify aptamers with polymerization-inhibitory properties. Two noncompeting aptamers, DE3A and OX3B, were found to bind hemoglobin, significantly increase the delay time, and reduce the rate of polymerization of HbS. These modifiable, nuclease-resistant aptamers are potential new therapeutic agents for SCD.


Asunto(s)
Antidrepanocíticos/administración & dosificación , Antidrepanocíticos/química , Aptámeros de Nucleótidos/química , Hemoglobina Falciforme/química , Polimerizacion/efectos de los fármacos , Anemia de Células Falciformes/tratamiento farmacológico , Anemia de Células Falciformes/metabolismo , Antidrepanocíticos/síntesis química , Aptámeros de Nucleótidos/administración & dosificación , Aptámeros de Nucleótidos/síntesis química , Sistema Libre de Células , ADN Complementario , Nucleótidos de Desoxicitosina/química , Nucleótidos de Desoxiuracil/química , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas/métodos , Humanos , Pirimidinas/química , Técnica SELEX de Producción de Aptámeros/métodos
5.
PLoS One ; 11(10): e0164288, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27755560

RESUMEN

Plasminogen activator inhibitor-1 (PAI-1) is elevated in various cancers, where it has been shown to effect cell migration and invasion and angiogenesis. While, PAI-1 is a secreted protein, its intercellular levels are increased in cancer cells. Consequently, intracellular PAI-1 could contribute to cancer progression. While various small molecule inhibitors of PAI-1 are currently being investigated, none specifically target intracellular PAI-1. A class of inhibitors, termed aptamers, has been used effectively in several clinical applications. We previously generated RNA aptamers that target PAI-1 and demonstrated their ability to inhibit extracellular PAI-1. In the current study we explored the effect of these aptamers on intracellular PAI-1. We transiently transfected the PAI-1 specific aptamers into both MDA-MB-231 human breast cancer cells, and human umbilical vein endothelial cells (HUVECs) and studied their effects on cell migration, invasion and angiogenesis. Aptamer expressing MDA-MB-231 cells exhibited a decrease in cell migration and invasion. Additionally, intracellular PAI-1 and urokinase plasminogen activator (uPA) protein levels decreased, while the PAI-1/uPA complex increased. Moreover, a significant decrease in endothelial tube formation in HUVECs transfected with the aptamers was observed. In contrast, conditioned media from aptamer transfected MDA-MB-231 cells displayed a slight pro-angiogenic effect. Collectively, our study shows that expressing functional aptamers inside breast and endothelial cells is feasible and may exhibit therapeutic potential.


Asunto(s)
Aptámeros de Nucleótidos/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Aptámeros de Nucleótidos/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular , Supervivencia Celular , Citocinas/análisis , Ensayo de Inmunoadsorción Enzimática , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Neovascularización Fisiológica , Inhibidor 1 de Activador Plasminogénico/química , Inhibidor 1 de Activador Plasminogénico/genética , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Vitronectina/metabolismo
6.
Oncotarget ; 6(32): 33534-53, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26392335

RESUMEN

The membrane-anchored serine proteases are a unique group of trypsin-like serine proteases that are tethered to the cell surface via transmembrane domains or glycosyl-phosphatidylinositol-anchors. Overexpressed in tumors, with pro-tumorigenic properties, they are attractive targets for protease-activated prodrug-like anti-tumor therapies. Here, we sought to engineer anthrax toxin protective antigen (PrAg), which is proteolytically activated on the cell surface by the proprotein convertase furin to instead be activated by tumor cell-expressed membrane-anchored serine proteases to function as a tumoricidal agent. PrAg's native activation sequence was mutated to a sequence derived from protein C inhibitor (PCI) that can be cleaved by membrane-anchored serine proteases, to generate the mutant protein PrAg-PCIS. PrAg-PCIS was resistant to furin cleavage in vitro, yet cytotoxic to multiple human tumor cell lines when combined with FP59, a chimeric anthrax toxin lethal factor-Pseudomonas exotoxin fusion protein. Molecular analyses showed that PrAg-PCIS can be cleaved in vitro by several serine proteases including the membrane-anchored serine protease testisin, and mediates increased killing of testisin-expressing tumor cells. Treatment with PrAg-PCIS also potently attenuated the growth of testisin-expressing xenograft tumors in mice. The data indicates PrAg can be engineered to target tumor cell-expressed membrane-anchored serine proteases to function as a potent tumoricidal agent.


Asunto(s)
Antígenos Bacterianos/farmacología , Toxinas Bacterianas/farmacología , Profármacos/farmacología , Serina Endopeptidasas/farmacología , Secuencia de Aminoácidos , Animales , Antígenos Bacterianos/genética , Antineoplásicos/farmacología , Toxinas Bacterianas/genética , Línea Celular Tumoral , Femenino , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/farmacología , Células HEK293 , Células HeLa , Humanos , Ratones , Ratones Desnudos , Ingeniería de Proteínas , Serina Endopeptidasas/genética , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Nucleic Acid Ther ; 24(4): 239-49, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24922319

RESUMEN

Plasminogen activator inhibitor-1 (PAI-1; SERPINE1) inhibits the plasminogen activators: tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA). Elevated levels of PAI-1 have been correlated with an increased risk for cardiovascular disease. Pharmacologically suppressing PAI-1 might prevent, or successfully treat PAI-1 related vascular diseases. This can potentially be accomplished by using small RNA molecules (aptamers). This study's goal is to develop RNA aptamers to a region of PAI-1 that will prevent the ability of PAI-1 to interact with the plasminogen activators. The aptamers were generated through a systematic evolution of ligands via exponential enrichment approach that ensures the creation of RNA molecules that bind to our target protein, PAI-1. In vitro assays were used to determine the effect of these aptamers on PAI-1's inhibitory activity. Three aptamers that bind to PAI-1 with affinities in the nanomolar range were isolated. The aptamer clones R10-4 and R10-2 inhibited PAI-1's antiproteolytic activity against tPA and disrupted PAI-1's ability to form a stable covalent complex with tPA. Increasing aptamer concentrations correlated positively with an increase in cleaved PAI-1. To the best of our knowledge, this is the first report of RNA molecules that inhibit the antiproteolytic activity of PAI-1.


Asunto(s)
Aptámeros de Nucleótidos/química , Inhibidor 1 de Activador Plasminogénico/química , Inhibidores de Proteasas/química , Activador de Tejido Plasminógeno/química , Aptámeros de Nucleótidos/síntesis química , Aptámeros de Nucleótidos/metabolismo , Humanos , Inhibidor 1 de Activador Plasminogénico/metabolismo , Inhibidores de Proteasas/metabolismo , Unión Proteica , Proteolisis , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Técnica SELEX de Producción de Aptámeros , Activador de Tejido Plasminógeno/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/química , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
8.
Expert Opin Ther Pat ; 23(7): 801-15, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23521527

RESUMEN

INTRODUCTION: Plasminogen activator inhibitor-1 (PAI-1), the serine protease inhibitor (serpin), binds to and inhibits the plasminogen activators-tissue-type plasminogen activator (tPA) and the urokinase-type plasminogen activator (uPA). This results in both a decrease in plasmin production and a decrease in the dissolution of fibrin clots. Elevated levels of PAI-1 are correlated with an increased risk for cardiovascular disease and have been linked to obesity and metabolic syndrome. Consequently, the pharmacological suppression of PAI-1 might prevent or treat vascular disease. AREAS COVERED: This article provides an overview of the patenting activity on PAI-1 inhibitors. Patents filed by pharmaceutical companies or individual research groups are described, and the biological and biochemical evaluation of the inhibitors, including in vitro and in vivo studies, is discussed. An overview of patents pertaining to using these inhibitors for treating various diseases is also included. EXPERT OPINION: Although there is still no PAI-1 inhibitor being evaluated in a clinical setting or approved for human therapy, research in this field has progressed, and promising new compounds have been designed. Most research has focused on improving the pharmacological profile of these compounds, which will hopefully allow them to proceed to clinical studies. Despite the need for further testing and research, the potential use of PAI-1 inhibitors for treating cardiovascular disease appears quite promising.


Asunto(s)
Enfermedades Cardiovasculares/tratamiento farmacológico , Diseño de Fármacos , Inhibidor 1 de Activador Plasminogénico/metabolismo , Animales , Enfermedades Cardiovasculares/fisiopatología , Fibrinolisina/metabolismo , Humanos , Patentes como Asunto
9.
Nucleic Acid Ther ; 21(6): 373-81, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22103403

RESUMEN

The serine protease inhibitor (serpin) plasminogen activator inhibitor-1 (PAI-1) is associated with the pathophysiology of several diseases, including cancer and cardiovascular disease. The extracellular matrix protein vitronectin increases at sites of vessel injury and is also present in fibrin clots. Integrins present on the cell surface bind to vitronectin and anchor the cell to the extracellular matrix. However, the binding of PAI-1 to vitronectin prevents this interaction, thereby decreasing both cell adhesion and migration. We previously developed PAI-1-specific RNA aptamers that bind to (or in the vicinity of) the vitronectin binding site of PAI-1. These aptamers prevented cancer cells from detaching from vitronectin in the presence of PAI-1, resulting in an increase in cell adhesion. In the current study, we used in vitro assays to investigate the effects that these aptamers have on human aortic smooth muscle cell (HASMC) and human umbilical vein endothelial cell (HUVEC) migration, adhesion, and proliferation. The PAI-1-specific aptamers (SM20 and WT15) increased attachment of HASMCs and HUVECs to vitronectin in the presence of PAI-1 in a dose-dependent manner. Whereas PAI-1 significantly inhibited cell migration through its interaction with vitronectin, both SM20 and WT15 restored cell migration. The PAI-1 vitronectin binding mutant (PAI-1AK) did not facilitate cell detachment or have an effect on cell migration. The effect on cell proliferation was minimal. Additionally, both SM20 and WT15 promoted tube formation on matrigel that was supplemented with vitronectin, thereby reversing the PAI-1's inhibition of tube formation. Collectively, results from this study show that SM20 and WT15 bind to the PAI-1's vitronectin binding site and interfere with its effect on cell migration, adhesion, and tube formation. By promoting smooth muscle and endothelial cell migration, these aptamers can potentially eliminate the adverse effects of elevated PAI-1 levels in the pathogenesis of vascular disease.


Asunto(s)
Aptámeros de Nucleótidos/administración & dosificación , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Inhibidor 1 de Activador Plasminogénico/metabolismo , Unión Proteica/efectos de los fármacos , Vitronectina/metabolismo , Aorta/citología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Venas Umbilicales/citología
10.
Semin Thromb Hemost ; 37(4): 339-48, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21805439

RESUMEN

Heparin cofactor II (HCII) is a serine protease inhibitor (serpin) found in high concentrations in human plasma. Despite its discovery >30 years ago, its physiological function is still poorly understood. It is known to inhibit thrombin, the predominant coagulation protease, and HCII-thrombin complexes have been found in plasma, yet it is thought to contribute little to normal hemostasis. However, thrombin has several other physiological functions, and therefore many biological roles for HCII need consideration. The unique structure and mechanism of action of HCII have helped guide our understanding of HCII. In particular, HCII binds many glycosaminoglycans (GAGs) such as heparin and heparin sulfate as well as several different polyanions to enhance its inhibition of thrombin. Distinctly, HCII is able to use the GAG dermatan sulfate for accelerated thrombin inhibition. Dermatan sulfate is found in high concentrations in the walls of blood vessels as well as in placental tissue. This knowledge has led to research indicating that HCII may play a protective role in atherosclerosis and placental thrombosis. Additionally, pharmaceuticals are being developed that use the dermatan sulfate activation of HCII for anticoagulation. Although much research is still needed to fully understand HCII, this humble protein may have significant impact in our medical future. This article reviews the laboratory history, protein characteristics, structure-activity relationships, protease inhibition, physiological function, and medical relevance of HCII in hopes of regenerating interest in this sometimes forgotten serpin.


Asunto(s)
Cofactor II de Heparina/fisiología , Animales , Cofactor II de Heparina/química , Homeostasis/fisiología , Humanos , Enfermedades Vasculares/sangre , Enfermedades Vasculares/tratamiento farmacológico
11.
PLoS One ; 5(5): e10846, 2010 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-20520766

RESUMEN

BACKGROUND: Dimeric naphthoquinones (BiQ) were originally synthesized as a new class of HIV integrase inhibitors but have shown integrase-independent cytotoxicity in acute lymphoblastic leukemia cell lines suggesting their use as potential anti-neoplastic agents. The mechanism of this cytotoxicity is unknown. In order to gain insight into the mode of action of binaphthoquinones we performed a systematic high-throughput screen in a yeast isogenic deletion mutant array for enhanced or suppressed growth in the presence of binaphthoquinones. METHODOLOGY/PRINCIPAL FINDINGS: Exposure of wild type yeast strains to various BiQs demonstrated inhibition of yeast growth with IC(50)s in the microM range. Drug sensitivity and resistance screens were performed by exposing arrays of a haploid yeast deletion mutant library to BiQs at concentrations near their IC(50). Sensitivity screens identified yeast with deletions affecting mitochondrial function and cellular respiration as having increased sensitivity to BiQs. Corresponding to this, wild type yeast grown in the absence of a fermentable carbon source were particularly sensitive to BiQs, and treatment with BiQs was shown to disrupt the mitochondrial membrane potential and lead to the generation of reactive oxygen species (ROS). Furthermore, baseline ROS production in BiQ sensitive mutant strains was increased compared to wild type and could be further augmented by the presence of BiQ. Screens for resistance to BiQ action identified the mitochondrial external NAD(P)H dehydrogenase, NDE1, as critical to BiQ toxicity and over-expression of this gene resulted in increased ROS production and increased sensitivity of wild type yeast to BiQ. CONCLUSIONS/SIGNIFICANCE: In yeast, binaphthoquinone cytotoxicity is likely mediated through NAD(P)H:quonine oxidoreductases leading to ROS production and dysfunctional mitochondria. Further studies are required to validate this mechanism in mammalian cells.


Asunto(s)
Pruebas Genéticas , Viabilidad Microbiana/efectos de los fármacos , Naftoquinonas/toxicidad , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/efectos de los fármacos , Aerobiosis/efectos de los fármacos , Medios de Cultivo , Fermentación/efectos de los fármacos , Genoma Fúngico/genética , Isomerismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Naftoquinonas/química , Especies Reactivas de Oxígeno/metabolismo , Reproducibilidad de los Resultados , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
12.
Biochim Biophys Acta ; 1800(6): 580-90, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20230872

RESUMEN

BACKGROUND: Protein C inhibitor (PCI) is a plasma serine protease inhibitor (serpin) that regulates several serine proteases in coagulation including thrombin and activated protein C. However, the physiological role of PCI remains under investigation. The cysteine protease, cathepsin L, has a role in many physiological processes including cardiovascular diseases, blood vessel remodeling, and cancer. METHODS AND RESULTS: We found that PCI inhibits cathepsin L with an inhibition rate (k(2)) of 3.0x10(5)M(-)(1)s(-)(1). Whereas, the PCI P1 mutant (R354A) inhibits cathepsin L at rates similar to wild-type PCI, mutating the P2 residue results in a slight decrease in the rate of inhibition. We then assessed the effect of PCI and cathepsin L on the migration of human breast cancer (MDA-MB-231) cells. Cathepsin L was expressed in both the cell lysates and conditioned media of MDA-MB-231 cells. Wound-induced and transwell migration of MDA-MB-231 cells was inhibited by exogenously administered wtPCI and PCI P1 but not PCI P14 mutant. In addition, migration of MDA-MB-231 cells expressing wtPCI was significantly decreased compared to non-expressing MDA-MB-231 cells or MDA-MB-231 cells expressing the PCI P14 mutant. Downregulation of cathepsin L by either a specific cathepsin L inhibitor or siRNA technology also resulted in a decrease in the migration of MDA-MB-231 cells. CONCLUSIONS: Overall, our data show that PCI regulates tumor cell migration partly by inhibiting cathepsin L. GENERAL SIGNIFICANCE: Consequently, inhibiting cathepsin L by serpins like PCI may be a new pathway of regulating hemostasis, cardiovascular and metastatic diseases.


Asunto(s)
Neoplasias de la Mama/patología , Catepsina L/metabolismo , Metástasis de la Neoplasia , Inhibidor de Proteína C/farmacología , Secuencia de Bases , Neoplasias de la Mama/enzimología , Línea Celular Tumoral , Cartilla de ADN , Técnica del Anticuerpo Fluorescente , Humanos , Mutagénesis Sitio-Dirigida
13.
Oligonucleotides ; 19(2): 117-28, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19284310

RESUMEN

The serine protease inhibitor plasminogen activator inhibitor-1 (PAI-1) is increased in several cancers, including breast, where it is associated with a poor outcome. Metastatic breast cancer has a dismal prognosis, as evidenced by treatment goals that are no longer curative but are largely palliative in nature. PAI-1 competes with integrins and the urokinase plasminogen activator receptor on the surface of breast cancer cells for binding to vitronectin. This results in the detachment of tumor cells from the extracellular matrix, which is critical to the metastatic process. For this reason, we sought to isolate RNA aptamers that disrupt the interaction between PAI-1 and vitronectin. Through utilization of combinatorial chemistry techniques, aptamers have been selected that bind to PAI-1 with high affinity and specificity. We identified two aptamers, WT-15 and SM-20, that disrupt the interactions between PAI-1 and heparin, as well as PAI-1 and vitronectin, without affecting the antiprotease activity of PAI-1. Furthermore, SM-20 prevented the detachment of breast cancer cells (MDA-MB-231) from vitronectin in the presence of PAI-1, resulting in an increase in cellular adhesion. Therefore, the PAI-1 aptamer SM-20 demonstrates therapeutic potential as an antimetastatic agent and could possibly be used as an adjuvant to traditional chemotherapy for breast cancer.


Asunto(s)
Aptámeros de Nucleótidos/farmacología , Neoplasias de la Mama/terapia , Inhibidor 1 de Activador Plasminogénico/metabolismo , ARN/farmacología , Vitronectina/metabolismo , Aptámeros de Nucleótidos/química , Neoplasias de la Mama/patología , Adhesión Celular/efectos de los fármacos , Heparina/metabolismo , Humanos , Metástasis de la Neoplasia , ARN/química , Técnica SELEX de Producción de Aptámeros , Trombina/metabolismo
14.
Biochim Biophys Acta ; 1748(1): 57-65, 2005 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15752693

RESUMEN

The serine protease inhibitor (serpin) protein C inhibitor (PCI; also named plasminogen activator inhibitor-3) regulates serine proteases in hemostasis, fibrinolysis, and reproduction. The biochemical activity of PCI is not fully defined partly due to the lack of a convenient expression system for active rPCI. Using pET-15b plasmid, Ni(2+)-chelate and heparin-Sepharose affinity chromatography steps, we describe here the expression, purification and characterization of wild-type recombinant (wt-rPCI) and two inactive mutants, R354A (P1 residue) and T341R (P14 residue), expressed in Escherichia coli. Wild-type rPCI, but not the two mutants, formed a stable bimolecular complex with thrombin, activated protein C and urokinase. In the absence of heparin, wt-rPCI-thrombin, -activated protein C, and -urokinase inhibition rates were 56.7, 3.4, and 2.3 x 10(4) M(-1) min(-1), respectively, and the inhibition rates were accelerated 25-, 71-, and 265-fold in the presence of 10 mug/mL heparin for each respective inhibition reaction. The stoichiometry of inhibition (SI) for wt-rPCI-thrombin was 2.0, which is comparable to plasma-derived PCI. The present report describes for the first time the expression and characterization of recombinant PCI in a bacterial expression system and demonstrates the feasibility of using this system to obtain adequate amounts of biologically active rPCI for future structure-function studies.


Asunto(s)
Escherichia coli/metabolismo , Inhibidor de Proteína C/metabolismo , Inhibidores de Serina Proteinasa/metabolismo , Secuencia de Aminoácidos , Escherichia coli/genética , Heparina/metabolismo , Humanos , Datos de Secuencia Molecular , Mutación , Proteína C/antagonistas & inhibidores , Proteína C/metabolismo , Inhibidor de Proteína C/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Inhibidores de Serina Proteinasa/genética , Trombina/antagonistas & inhibidores , Trombina/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
15.
J Biol Chem ; 279(41): 43237-44, 2004 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-15292227

RESUMEN

We used 55 Ala-scanned recombinant thrombin molecules to define residues important for inhibition by the serine protease inhibitor (serpin) heparin cofactor II (HCII) in the absence and presence of glycosaminoglycans. We verified the importance of numerous basic residues in anion-binding exosite-1 (exosite-1) and found 4 additional residues, Gln24, Lys65, His66, and Tyr71 (using the thrombin numbering system), that were resistant to HCII inhibition with and without glycosaminoglycans. Inhibition rate constants for these exosite-1 (Q24A, K65A, H66A, Y71A) thrombin mutants (0.02-0.38 x 10(8) m(-1) min(-1) for HCII-heparin when compared with 2.36 x 10(8) m(-1) min(-1) with wild-type thrombin and 0.03-0.53 x 10(8) m(-1) min(-1) for HCII-dermatan sulfate when compared with 5.23 x 10(8) m(-1) min(-1) with wild-type thrombin) confirmed that the structural integrity of thrombin exosite-1 is critical for optimal HCII-thrombin interactions in the presence of glycosaminoglycans. However, our results are also consistent for HCII-glycosaminoglycan-thrombin ternary complex formation. Ten residues surrounding the active site of thrombin were implicated in HCII interactions. Four mutants (Asp51, Lys52, Lys145/Thr147/Trp148, Asp234) showed normal increased rates of inhibition by HCII-glycosaminoglycans, whereas four mutants (Trp50, Glu202, Glu229, Arg233) remained resistant to inhibition by HCII with glycosaminoglycans. Using 11 exosite-2 thrombin mutants with 20 different mutated residues, we saw no major perturbations of HCII-glycosaminoglycan inhibition reactions. Collectively, our results support a "double bridge" mechanism for HCII inhibition of thrombin in the presence of glycosaminoglycans, which relies in part on ternary complex formation but is primarily dominated by an allosteric process involving contact of the "hirudin-like" domain of HCII with thrombin exosite-1.


Asunto(s)
Cofactor II de Heparina/química , Trombina/química , Alanina/química , Sitio Alostérico , Sitios de Unión , Dermatán Sulfato/química , Relación Dosis-Respuesta a Droga , Glutamina/química , Glicosaminoglicanos/química , Histidina/química , Humanos , Cinética , Lisina/química , Modelos Moleculares , Mutagénesis , Mutación , Unión Proteica , Relación Estructura-Actividad , Trombina/metabolismo , Factores de Tiempo , Tirosina/química
16.
FEBS Lett ; 568(1-3): 10-4, 2004 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-15196911

RESUMEN

We studied the RNA aptamer Toggle-25/thrombin interaction during inhibition by antithrombin (AT), heparin cofactor II (HCII) and protein C inhibitor (PCI). Thrombin inhibition was reduced 3-fold by Toggle-25 for AT and HCII, but it was slightly enhanced for PCI. In the presence of glycosaminoglycans, AT and PCI had significantly reduced thrombin inhibition with Toggle-25, but it was only reduced 3-fold for HCII. This suggested that the primary effect of aptamer binding was through the heparin-binding site of thrombin, anion-binding exosite-2 (exosite-2). We localized the Toggle-25 binding site to Arg 98, Glu 169, Lys 174, Asp 175, Arg 245, and Lys 248 of exosite-2. We conclude that a RNA aptamer to thrombin exosite-2 might provide an effective clinical reagent to control heparin's anticoagulant action.


Asunto(s)
Antitrombinas/metabolismo , Cofactor II de Heparina/metabolismo , Heparina/metabolismo , Inhibidor de Proteína C/metabolismo , ARN/metabolismo , Trombina/metabolismo , Aniones , Secuencia de Bases , Sitios de Unión , Glicosaminoglicanos/metabolismo , Humanos , Modelos Moleculares , ARN/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...