Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Invest New Drugs ; 2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38789849

RESUMEN

Worldwide, pancreatic cancer (PC) is a major health problem and almost 0.5 million people were diagnosed with PC in 2020. In the United States, more than 64,000 adults will be diagnosed with PC in 2023. PC is highly resistant to currently available treatments and standard of care chemotherapies cause serious side effects. Most PC patients are resistant to clinical therapies. Combination therapy has showed superior efficacy over single-agent treatment. However, most therapy has failed to show a significant improvement in overall survival due to treatment-related toxicity. Developing efficacious clinically useful PC therapies remains a challenge. Herein, we show the efficacy of an innovative pathway modulator, p53-Activator Wnt Inhibitor-2 (PAWI-2) against tumors arising from human pancreatic cancer stem cells (i.e., hPCSCs, FGß3 cells). PAWI-2 is a potent inhibitor of tumor growth. In the present study, we showed PAWI-2 potently inhibited growth of tumors from hPCSCs in orthopic xenograft models of both male and female mice. PAWI-2 worked in a non-toxic manner to inhibit tumors. Compared to vehicle-treated animals, PAWI-2 modulated molecular regulators of tumors. Anti-cancer results showed PAWI-2 in vivo efficacy could be correlated to in vitro potency to inhibit FGß3 cells. PAWI-2 represents a safe, new approach to combat PC.

2.
Invest New Drugs ; 39(1): 131-141, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32915418

RESUMEN

Today, pancreatic cancer (PC) is a major health problem in the United States. It remains a challenge to develop efficacious clinically useful PC therapies. New avenues, based on translational approaches and innovative validated biomarkers could be a preclinical option to evaluate PC drug candidates or drug combinations before clinical trials. Herein, we describe evaluation of combination therapies by incorporating a novel pathway modulator, p53-Activator Wnt Inhibitor-2 (PAWI-2) with other FDA-approved cancer drugs that have been used in PC clinical trials. PAWI-2 is a potent inhibitor of drug-resistant PC cells that has been shown to selectively ameliorate human pancreatic cancer stem cells (i.e., hPCSCs, FGß3 cells). In the present study, we showed PAWI-2 produced therapeutic synergism with certain types of anti-cancer drugs. These drugs themselves oftentimes do not ameliorate PC cells (especially PCSCs) due to high levels of drug-resistance. PAWI-2 has the ability to rescue the potency of drugs (i.e., erlotinib, trametinib) and inhibit PC cell growth. Key molecular regulators of PAWI-2 could be used to predict synergistic/antagonistic effects between PAWI-2 and other anti-cancer drugs. Anti-cancer results showed potency could be quite accurately correlated to phosphorylation of optineurin (OPTN) in PC cells. Synergism/antagonism was also associated with inhibition of PCSC marker SOX2 that was observed in FGß3 cells. Synergism broadens the potential use of PAWI-2 as an adjunct chemotherapy in patients with PC that have developed resistance to first-line targeted therapies or chemotherapies.


Asunto(s)
Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Pancreáticas/patología , Quinoxalinas/farmacología , Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Humanos , Proteínas de Transporte de Membrana/efectos de los fármacos , Células Madre Neoplásicas/efectos de los fármacos , Quinoxalinas/administración & dosificación , Factores de Transcripción SOXB1/efectos de los fármacos
3.
Bioorg Med Chem Lett ; 46: 128162, 2021 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-34062251

RESUMEN

In the United States, approximately one million individuals are hospitalized every year for arrhythmias, making arrhythmias one of the top causes of healthcare expenditures. Mexiletine is currently used as an antiarrhythmic drug but has limitations. The purpose of this work was to use normal and Long QT syndrome Type 3 (LQTS3) patient-derived human induced pluripotent stem cell (iPSC)-derived cardiomyocytes to identify an analog of mexiletine with superior drug-like properties. Compared to racemic mexiletine, medicinal chemistry optimization of substituted racemic pyridyl phenyl mexiletine analogs resulted in a more potent sodium channel inhibitor with greater selectivity for the sodium over the potassium channel and for late over peak sodium current.


Asunto(s)
Trastorno del Sistema de Conducción Cardíaco/patología , Células Madre Pluripotentes Inducidas/química , Síndrome de QT Prolongado/patología , Mexiletine/farmacología , Miocitos Cardíacos/patología , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Piridinas/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Mexiletine/química , Estructura Molecular , Piridinas/química , Relación Estructura-Actividad
4.
Drug Metab Dispos ; 48(2): 106-115, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31727673

RESUMEN

Oxycodone is used as a potent analgesic medication. Oxycodone is extensively metabolized. To fully describe its metabolism, the oxygenation of oxycodone to oxycodone N-oxide was investigated in hepatic preparations. The hypothesis tested was that oxycodone N-oxygenation was enzymatic and the amount of N-oxide detected was a consequence of both oxygenation and retro-reduction. Methods for testing the hypothesis included both in vitro and in vivo studies. Results indicated that oxycodone was N-oxygenated by the flavin-containing monooxygenase. Oxycodone N-oxide is chemically quite stable but in the presence of hepatic preparations and NADPH was retro-reduced to its parent compound oxycodone. Subsequently, oxycodone was metabolized to other metabolites including noroxycodone, noroxymorphone, and oxymorphone via cytochrome P-450. Retro-reduction of oxycodone N-oxide to oxycodone was facilitated by quinone reductase, aldehyde oxidase, and hemoglobin but not to a great extent by cytochrome P-450 or the flavin-containing monooxygenase. To confirm the in vitro observations, oxycodone was administered to rats and humans. In good agreement with in vitro results, substantial oxycodone N-oxide was observed in urine after oxycodone administration to rats and humans. Administration of oxycodone N-oxide to rats showed substantial amount of recovered oxycodone N-oxide. In vivo, noroxycodone was formed as a major rat urinary metabolite from oxycodone N-oxide presumably after retro-reduction to oxycodone and oxidative N-demethylation. To a lesser extent, oxycodone, noroxymorphone, and oxymorphone were observed as urinary metabolites. SIGNIFICANCE STATEMENT: This manuscript describes the N-oxygenation of oxycodone in vitro as well as in small animals and humans. A new metabolite was quantified as oxycodone N-oxide. Oxycodone N-oxide undergoes extensive retro-reduction to oxycodone. This re-establishes the metabolic profile of oxycodone and introduces new concepts about a metabolic futile cycle related to oxycodone metabolism.


Asunto(s)
Óxidos/metabolismo , Oxicodona/metabolismo , Analgésicos Opioides/metabolismo , Animales , Sistema Enzimático del Citocromo P-450/metabolismo , Femenino , Hemoglobinas/metabolismo , Humanos , Masculino , Oxigenasas de Función Mixta/metabolismo , Morfinanos/metabolismo , NADP/metabolismo , Oximorfona/metabolismo , Ratas
5.
J Pharmacol Exp Ther ; 371(3): 703-712, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31582422

RESUMEN

Prostate cancer (PCa) is the second leading cause of cancer-related death for men in the United States. Approximately 35% of PCa recurs and is often transformed to castration-resistant prostate cancer (CRPCa), the most deadly and aggressive form of PCa. However, the CRPCa standard-of-care treatment (enzalutamide with abiraterone) usually has limited efficacy. Herein, we report a novel molecule (PAWI-2) that inhibits cellular proliferation of androgen-sensitive and androgen-insensitive cells (LNCaP and PC-3, respectively). In vivo studies in a PC-3 xenograft model showed that PAWI-2 (20 mg/kg per day i.p., 21 days) inhibited tumor growth by 49% compared with vehicle-treated mice. PAWI-2 synergized currently clinically used enzalutamide in in vitro inhibition of PCa cell viability and resensitized inhibition of in vivo PC-3 tumor growth. Compared with vehicle-treated mice, PC-3 xenograft studies also showed that PAWI-2 (20 mg/kg per day i.p., 21 days) and enzalutamide (5 mg/kg per day i.p., 21 days) inhibited tumor growth by 63%. Synergism was mainly controlled by the imbalance of prosurvival factors (e.g., Bcl-2, Bcl-xL, Mcl-1) and antisurvival factors (e.g., Bax, Bak) induced by affecting mitochondrial membrane potential/mitochondria dynamics. Thus, PAWI-2 utilizes a distinct mechanism of action to inhibit PCa growth independently of androgen receptor signaling and overcomes enzalutamide-resistant CRPCa. SIGNIFICANCE STATEMENT: Castration-resistant prostate cancer (CRPCa) is the most aggressive human prostate cancer (PCa) but standard chemotherapies for CRPCa are largely ineffective. PAWI-2 potently inhibits PCa proliferation in vitro and in vivo regardless of androgen receptor status and uses a distinct mechanism of action. PAWI-2 has greater utility in treating CRPCa than standard-of-care therapy. PAWI-2 possesses promising therapeutic potency in low-dose combination therapy with a clinically used drug (e.g., enzalutamide). This study describes a new approach to address the overarching challenge in clinical treatment of CRPCa.


Asunto(s)
Antineoplásicos/farmacología , Feniltiohidantoína/análogos & derivados , Piperazinas/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Quinoxalinas/farmacología , Animales , Apoptosis/efectos de los fármacos , Aspartato Aminotransferasas/sangre , Benzamidas , Línea Celular Tumoral , Sinergismo Farmacológico , Humanos , Masculino , Ratones , Nitrilos , Feniltiohidantoína/farmacología , Neoplasias de la Próstata/patología , Proteína p53 Supresora de Tumor/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Anal Chem ; 90(1): 974-979, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29172437

RESUMEN

Toxicity from acute exposure to nerve agents and organophosphorus toxicants is due to irreversible inhibition of acetylcholinesterase (AChE) in the nervous system. AChE in red blood cells is a surrogate for AChE in the nervous system. Previously we developed an immunopurification method to enrich red blood cell AChE (RBC AChE) as a biomarker of exposure. The goal of the present work was to provide an alternative RBC AChE enrichment strategy, by binding RBC AChE to Hupresin affinity gel. AChE was solubilized from frozen RBC by addition of 1% Triton X-100. Insoluble debris was removed by centrifugation. The red, but not viscous, RBC AChE solution was loaded on a Hupresin affinity column. Hemoglobin and other proteins were washed off with 3 M NaCl, while retaining AChE bound to Hupresin. Denatured AChE was eluted with 1% trifluoroacetic acid. The same protocol was used for 20 mL of RBC AChE inhibited with a soman model compound. The acid denatured protein was digested with pepsin and analyzed by liquid chromatography tandem mass spectrometry on a 6600 Triple-TOF mass spectrometer. A targeted method identified the aged soman adduct on serine 203 in peptide FGESAGAAS. It was concluded that Hupresin can be used to enrich soman-inhibited AChE solubilized from 8 mL of frozen human erythrocytes, yielding a quantity sufficient for detecting soman exposure.


Asunto(s)
Acetilcolinesterasa/análisis , Cromatografía de Afinidad/métodos , Agentes Nerviosos/análisis , Soman/análisis , Acetilcolinesterasa/química , Cromatografía de Afinidad/instrumentación , Pruebas de Enzimas , Eritrocitos/enzimología , Humanos , Agentes Nerviosos/química , Soman/química
7.
Bioorg Med Chem Lett ; 28(20): 3363-3367, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30201292

RESUMEN

The Notch signaling pathway is involved in cell proliferation and differentiation, and has been recognized as an active pathway in regenerating tissue and cancerous cells. Notch signaling inhibition is considered a viable approach to the treatment of a variety of conditions including colorectal cancer, pancreatic cancer, breast cancer and metastatic melanoma. The discovery that the b-annulated dihydropyridine FLI-06 (1) is an inhibitor of the Notch pathway with an EC50 ≈ 2.5 µM prompted us to screen a library of related analogs. After structure activity studies were conducted, racemic compound 7 was identified with an EC50 = 0.36 µM. Synthesis of individual enantiomers provided (+)-7 enantiomer with an EC50 = 0.13 µM, or about 20-fold the potency of 1.


Asunto(s)
Antineoplásicos/farmacología , Dihidropiridinas/farmacología , Receptor Notch1/antagonistas & inhibidores , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Dihidropiridinas/síntesis química , Dihidropiridinas/química , Células HCT116 , Humanos , Estructura Molecular , Quinolinas/síntesis química , Quinolinas/química , Quinolinas/farmacología , Estereoisomerismo
8.
Bioorg Med Chem ; 26(15): 4441-4451, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-30075999

RESUMEN

For adult women in the United States, breast cancer is the most prevalent form of cancer. Compounds that target dysregulated signal transduction can be efficacious anti-cancer therapies. A prominent signaling pathway frequently dysregulated in breast cancer cells is the Wingless-related integration site (Wnt) pathway. The purpose of the work was to optimize a "hit" from a screening campaign. 76,000 compounds were tested in a Wnt transcription assay and revealed potent and reproducible "hit," compound 1. Medicinal chemistry optimization of 1 led to more potent and drug-like molecules, 19, 24 and 25 (i.e., Wnt pathway IC50 values = 11, 18 and 7 nM, respectively). The principal results showed compounds 19, 24 and 25 were potent anti-proliferative agents in breast cancer cell lines, MCF-7 (i.e., IC50 values = 10, 7 and 4 nM, respectively) and MDA-MB 231 (i.e., IC50 values = 13, 13 and 16 nM, respectively). Compound 19 synergized anti-proliferation with chemotherapeutic Doxorubicin in vitro. A major conclusion was that compound 19 enhanced anti-proliferation of Doxorubicin in vitro and in a xenograft animal model of breast cancer.


Asunto(s)
Antineoplásicos/química , Sulfonamidas/química , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Doxorrubicina/farmacología , Sinergismo Farmacológico , Femenino , Humanos , Ratones , Ratones Desnudos , Relación Estructura-Actividad , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Transcripción Genética/efectos de los fármacos , Trasplante Heterólogo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
9.
Chem Res Toxicol ; 30(10): 1897-1910, 2017 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-28892361

RESUMEN

Nerve agents and organophosphorus pesticides make a covalent bond with the active site serine of acetylcholinesterase (AChE), resulting in inhibition of AChE activity and toxic symptoms. AChE in red blood cells (RBCs) serves as a surrogate for AChE in the nervous system. Mass spectrometry analysis of adducts on RBC AChE could provide evidence of exposure. Our goal was to develop a method of immunopurifying human RBC AChE in quantities adequate for detecting exposure by mass spectrometry. For this purpose, we immobilized 3 commercially available anti-human acetylcholinesterase monoclonal antibodies (AE-1, AE-2, and HR2) plus 3 new monoclonal antibodies. The monoclonal antibodies were characterized for binding affinity, epitope mapping by pairing analysis, and nucleotide and amino acid sequences. AChE was solubilized from frozen RBCs with 1% (v/v) Triton X-100. A 16 mL sample containing 5.8 µg of RBC AChE was treated with a quantity of soman model compound that inhibited 50% of the AChE activity. Native and soman-inhibited RBC AChE samples were immunopurified on antibody-Sepharose beads. The immunopurified RBC AChE was digested with pepsin and analyzed by liquid chromatography tandem mass spectrometry on a 6600 Triple-TOF mass spectrometer. The aged soman-modified PheGlyGluSerAlaGlyAlaAlaSer (FGESAGAAS) peptide was detected using a targeted analysis method. It was concluded that all 6 monoclonal antibodies could be used to immunopurify RBC AChE and that exposure to nerve agents could be detected as adducts on the active site serine of RBC AChE.


Asunto(s)
Acetilcolinesterasa/aislamiento & purificación , Eritrocitos/enzimología , Inmunoprecipitación , Agentes Nerviosos/análisis , Acetilcolinesterasa/inmunología , Acetilcolinesterasa/metabolismo , Humanos , Espectrometría de Masas
10.
Bioorg Med Chem ; 23(17): 5282-92, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26278027

RESUMEN

Cardiomyopathy is the leading cause of death worldwide. Despite progress in medical treatments, heart transplantation is one of the only current options for those with infarcted heart muscle. Stem cell differentiation technology may afford cell-based therapeutics that may lead to the generation of new, healthy heart muscle cells from undifferentiated stem cells. Our approach is to use small molecules to stimulate stem cell differentiation. Herein, we describe a novel class of 1,5-disubstituted benzimidazoles that induce differentiation of stem cells into cardiac cells. We report on the evaluation in vitro for cardiomyocyte differentiation and describe structure-activity relationship results that led to molecules with drug-like properties. The results of this study show the promise of small molecules to direct stem cell lineage commitment, to probe signaling pathways and to develop compounds for the stimulation of stem cells to repair damaged heart tissue.


Asunto(s)
Bencimidazoles/química , Bencimidazoles/farmacología , Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/efectos de los fármacos , Miocitos Cardíacos/citología , Animales , Células Cultivadas , Ratones , Miocitos Cardíacos/efectos de los fármacos , Relación Estructura-Actividad
11.
Biochemistry ; 53(27): 4476-87, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24902043

RESUMEN

Human butyrylcholinesterase (hBChE) is currently being developed as a detoxication enzyme for stoichiometric binding and/or catalytic hydrolysis of organophosphates. Herein, we describe the use of a molecular evolution method to develop novel hBChE variants with increased resistance to stereochemically defined nerve agent model compounds of soman, sarin, and cyclosarin. Novel hBChE variants (Y332S, D340H, and Y332S/D340H) were identified with an increased resistance to nerve agent model compounds that retained robust intrinsic catalytic efficiency. Molecular dynamics simulations of these variants revealed insights into the mechanism by which these structural changes conferred nerve agent model compound resistance.


Asunto(s)
Butirilcolinesterasa/química , Sustancias para la Guerra Química/química , Compuestos Organofosforados/química , Sarín/química , Soman/química , Butirilcolinesterasa/genética , Butiriltiocolina/química , Sustancias para la Guerra Química/toxicidad , Inhibidores de la Colinesterasa/química , Evolución Molecular Dirigida , Humanos , Hidrólisis , Cinética , Simulación de Dinámica Molecular , Compuestos Organofosforados/toxicidad , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Sarín/toxicidad , Soman/toxicidad
12.
J Pharmacol Exp Ther ; 350(1): 171-80, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24817033

RESUMEN

A substituted aryl amide derivative of 6-naltrexamine--17-cyclopropylmethyl-3,14ß-dihydroxy-4,5α-epoxy-6ß-[(4'-trimethylfluoro)benzamido]morphinan-hydrochloride--(compound 5), previously shown to be a potent κ-opioid receptor antagonist, was used to characterize the physicochemical properties and efficacy to decrease alcohol self-administration in alcohol-preferring rats (P-rats) and binge-like P-rats. Previous studies showed that compounds closely related to compound 5 possessed favorable properties regarding penetration of the blood-brain barrier. Pharmacokinetic studies showed that compound 5 had acceptable bioavailability. In contrast to other κ-receptor antagonists, in particular norbinaltorphimine, compound 5 showed favorable drug-like properties. Based on these findings, further studies were done. Safety studies showed that compound 5 was not hepatotoxic at doses 200-fold greater than an efficacious dose. The effects of compound 5 or naltrexone on the hepatotoxicity of thiobenzamide were investigated. In contrast to naltrexone, which exacerbated thiobenzamide-mediated hepatotoxicity, compound 5 was observed to be hepatoprotective. Based on the physicochemical properties of compound 5, the compound was examined in rat animal models of alcohol self-administration. The inhibition of ethanol self-administration by compound 5 in alcohol-dependent and alcohol-nondependent P-rats trained to self-administer a 10% (w/v) ethanol solution, using operant techniques, showed very potent efficacy (i.e., estimated ED50 values of 4-5 µg/kg). In a binge-like P-rat animal model, inhibition of alcohol self-administration by compound 5 had an estimated ED50 value of 8 µg/kg. The results suggest that compound 5 is a potent drug-like κ-opioid receptor antagonist of utility in alcohol cessation medications development.


Asunto(s)
Etanol/administración & dosificación , Morfinanos/farmacología , Naltrexona/análogos & derivados , Antagonistas de Narcóticos/farmacología , Alanina Transaminasa/sangre , Alanina Transaminasa/efectos de los fármacos , Animales , Aspartato Aminotransferasas/sangre , Aspartato Aminotransferasas/efectos de los fármacos , Enfermedad Hepática Inducida por Sustancias y Drogas/sangre , Condicionamiento Operante/efectos de los fármacos , Masculino , Morfinanos/efectos adversos , Morfinanos/farmacocinética , Naltrexona/efectos adversos , Antagonistas de Narcóticos/efectos adversos , Antagonistas de Narcóticos/farmacocinética , Ratas , Ratas Endogámicas , Autoadministración , Tioamidas/antagonistas & inhibidores
13.
J Biochem Mol Toxicol ; 28(1): 23-31, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23943350

RESUMEN

A countermeasure that protects the brain from organophosphate toxicity is an unmet need. Few small molecule reactivators that can cross the blood brain barrier and reactivate brain acetyl cholinesterases have been reported. Herein, we describe preclinical investigations of a new class of amidine-oxime reactivator of cholinesterases with improved potency and blood brain barrier permeability. (Z)-N-((E)-1-(Dimethylamino)-2-(hydroxyimino)ethylidene)butan-1-aminium chloride, 1, is zwitterionic at physiological pH but possesses increased oxime nucleophilicity because of the adjacent amidine functionality. The amidine-oximes reported herein were observed to be nontoxic (up to 200 mg/kg in vivo) and are chemically and metabolically stable. The results presented herein show that uncharged amidine-oxime reactivators such as 1 can penetrate the blood brain barrier in animals and protect from the toxicity of nerve agent model compounds.


Asunto(s)
Inhibidores de la Colinesterasa/toxicidad , Reactivadores de la Colinesterasa/farmacología , Organofosfatos/toxicidad , Oximas/farmacología , Animales , Barrera Hematoencefálica , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Reactivadores de la Colinesterasa/farmacocinética , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Oximas/farmacocinética , Ratas , Ratas Sprague-Dawley
14.
J Pharmacol Exp Ther ; 344(2): 531-41, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23192655

RESUMEN

A major challenge in organophosphate (OP) research has been the identification and utilization of reliable biomarkers for the rapid, sensitive, and efficient detection of OP exposure. Although Tyr 411 OP adducts to human serum albumin (HSA) have been suggested to be one of the most robust biomarkers in the detection of OP exposure, the analysis of HSA-OP adduct detection has been limited to techniques using mass spectrometry. Herein, we describe the procurement of two monoclonal antibodies (mAb-HSA-GD and mAb-HSA-VX) that recognized the HSA Tyr 411 adduct of soman (GD) or S-[2-(diisopropylamino)ethyl]-O-ethyl methylphosphonothioate (VX), respectively, but did not recognize nonphosphonylated HSA. We showed that mAb-HSA-GD was able to detect the HSA Tyr 411 OP adduct at a low level (i.e., human blood plasma treated with 180 nM GD) that could not be detected by mass spectrometry. mAb-HSA-GD and mAb-HSA-VX showed an extremely low-level detection of GD adducted to HSA (on the order of picograms). mAb-HSA-GD could also detect serum albumin OP adducts in blood plasma samples from different animals administered GD, including rats, guinea pigs, and monkeys. The ability of the two antibodies to selectively recognize nerve agents adducted to serum albumin suggests that these antibodies could be used to identify biomarkers of OP exposure and provide a new biologic approach to detect OP exposure in animals.


Asunto(s)
Anticuerpos Monoclonales , Sustancias para la Guerra Química/metabolismo , Exposición a Riesgos Ambientales/análisis , Compuestos Organofosforados/metabolismo , Albúmina Sérica/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , Antígenos/química , Antígenos/inmunología , Biomarcadores/sangre , Línea Celular Tumoral , Sustancias para la Guerra Química/química , Cromatografía Líquida de Alta Presión , Ensayo de Inmunoadsorción Enzimática , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Intoxicación por Organofosfatos/sangre , Compuestos Organofosforados/química , Unión Proteica , Ratas , Ratas Sprague-Dawley , Albúmina Sérica/química , Espectrometría de Masa por Ionización de Electrospray , Tirosina/química , Tirosina/metabolismo
15.
J Pharmacol Exp Ther ; 343(3): 673-82, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22956723

RESUMEN

Human butyrylcholinesterase (hBChE) is currently being developed as a detoxication enzyme for the catalytic hydrolysis or stoichiometric binding of organophosphates (OPs). Previously, rationally designed hBChE mutants (G117H and E197Q) were reported in the literature and showed the feasibility of engineering OP hydrolytic functional activity into hBChE. However, the OP hydrolysis rate for G117H is too low for clinical utility. Additional OP-resistant hBChE variants with greater hydrolysis rates are needed as OP nerve-agent countermeasures for therapeutic utility. As described herein, a directed molecular evolution process was used to identify amino acid residues that contribute to OP-resistant functional activity of hBChE variants. In this article, we describe the development and validation of a novel method to identify hBChE variants with OP-resistant functional activity (decreased rate of OP inhibition). The method reported herein used an adenoviral protein expression system combined with a functional screening protocol of OP nerve-agent model compounds that have been shown to have functional properties similar to authentic OP nerve-agent compounds. The hBChE screening method was robust for transfection efficiency, library diversity, and reproducibility of positive signals. The screening approach not only identified the previously reported hBChE G117H variant, but also identified a series of additional hBChE variants, including hBChE G117N, G117R, E197C, and L125V, that exhibited OP-resistant functional activities not reported previously. The mammalian functional screening approach can serve as a cornerstone for further optimization and screening for OP-resistant hBChEs for potential therapeutic applications.


Asunto(s)
Bioensayo/métodos , Butirilcolinesterasa/química , Butirilcolinesterasa/genética , Sustancias para la Guerra Química/toxicidad , Mutación , Compuestos Organofosforados/toxicidad , Adenoviridae/genética , Animales , Biocatálisis , Butirilcolinesterasa/metabolismo , Células CHO , Dominio Catalítico , Técnicas de Cultivo de Célula , Sustancias para la Guerra Química/química , Clonación Molecular , Cricetinae , Evolución Molecular Dirigida , Diseño de Fármacos , Vectores Genéticos , Células HEK293 , Humanos , Hidrólisis , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Compuestos Organofosforados/química , Ingeniería de Proteínas , Transfección
16.
Protein Expr Purif ; 80(1): 22-7, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21802514

RESUMEN

Human butyrylcholinesterase (BChE) can scavenge and thereby provide protection against various toxic esters, including organophosphate-based chemical warfare agents and the recreational drug cocaine. It is currently being used in molecular evolution studies to generate novel enzymes with improved ability to hydrolyze toxic ester compounds. Currently, the most commonly used purification strategies for recombinant BChE enzymes involve using affinity resins based on small molecule interactions with the enzyme's substrate binding site. However, as BChE variants are discovered and developed, a generic purification protocol that is insensitive to amino acid substitutions is necessary. In the current manuscript, an expression vector encoding a C-terminal truncation and a His6-tag was designed for BChE and used to express recombinant "wild-type" enzyme and two variants (i.e., G117H BChE and G117H/E197Q BChE). All the three His6-tagged enzymes were successfully purified via metal-affinity columns using similar procedures with good recovery. Steady-state kinetic parameters were determined for each enzyme, and values were compared to those obtained with the corresponding non-truncated non-His6-tagged enzymes. Rates of inhibition by echothiophate, a model compound for organophosphate-based pesticides, and rates of oxime-mediated reactivation after inhibition with a nerve agent model compound were also determined for selected enzymes. Rates of spontaneous reactivation from ETP inhibition were determined for the G117H variants. In all instances examined, truncation of the C-terminus of BChE and introduction of a His6-tag had no significant effects on the observed kinetic parameters, making this a highly useful construct for in vitro characterization of wild-type and variant BChEs.


Asunto(s)
Butirilcolinesterasa/genética , Butirilcolinesterasa/aislamiento & purificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Sustitución de Aminoácidos , Butirilcolinesterasa/química , Butirilcolinesterasa/metabolismo , Línea Celular , Sustancias para la Guerra Química/metabolismo , Expresión Génica , Histidina/genética , Humanos , Oligopéptidos/genética , Compuestos Organofosforados/metabolismo , Plásmidos/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
17.
Br J Clin Pharmacol ; 71(4): 585-91, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21395651

RESUMEN

AIM: The aim of this study was to investigate intra- and inter-individual variations of functional metabolic capacity of flavin-containing mono-oxygenase (FMO) during childhood using trimethylamine N-oxygenation as a probe reaction. METHODS: Trimethylamine N-oxygenation functional activity and presence of FMO1 (fetal form), FMO3 (adult form), and FMO5 (endogenous form) were immunochemically determined and compared in human liver microsomes obtained from children at various ages. As a control, the same parameters were studied with recombinant FMO1, FMO3 and FMO5 proteins as enzyme sources. Developmental variation in functional metabolic capacity of FMO was estimated by measuring urinary trimethylamine and its N-oxide in several individuals at different ages and in a group of 77 subjects in childhood. RESULTS: There was a significant correlation between trimethylamine N-oxygenation functional activity and FMO3 expression levels in human liver microsomes (r= 0.71, P < 0.05, n= 9). Trimethylamine N-oxygenation was catalyzed largely by FMO3 and not by FMO1 or FMO5. On the basis of analysis of intra-individual observations and collective urine samples under daily dietary conditions it was possible that neonates or infants harbouring at least one non-inactive-allele of the FMO3 gene could have developmental FMO3 metabolic capacity in childhood. CONCLUSIONS: Developmental variations in functional metabolic capacity of FMO3 in childhood were shown both on the basis of in vivo phenotyping tests and in in vitro liver microsomal determinations.


Asunto(s)
Antioxidantes/metabolismo , Expresión Génica/genética , Microsomas Hepáticos/metabolismo , Oxigenasas/metabolismo , Factores de Edad , Desarrollo Infantil , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Errores Innatos del Metabolismo/metabolismo , Metilaminas/metabolismo , Metilaminas/orina , Oxigenasas/genética , Polimorfismo Genético/genética , Análisis de Regresión
18.
Pharmacol Res Perspect ; 9(4): e00828, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34327875

RESUMEN

Prolongation of the cardiac action potential (AP) and early after depolarizations (EADs) are electrical anomalies of cardiomyocytes that can lead to lethal arrhythmias and are potential liabilities for existing drugs and drug candidates in development. For example, long QT syndrome-3 (LQTS3) is caused by mutations in the Nav 1.5 sodium channel that debilitate channel inactivation and cause arrhythmias. We tested the hypothesis that a useful drug (i.e., mexiletine) with potential liabilities (i.e., potassium channel inhibition and adverse reactions) could be re-engineered by dynamic medicinal chemistry to afford a new drug candidate with greater efficacy and less toxicity. Human cardiomyocytes were generated from LQTS3 patient-derived induced pluripotent stem cells (hIPSCs) and normal hIPSCs to determine beneficial (on-target) and detrimental effects (off-target) of mexiletine and synthetic analogs, respectively. The approach combined "drug discovery" and "hit to lead" refinement and showed that iterations of medicinal chemistry and physiological testing afforded optimized compound 22. Compared to mexiletine, compound 22 showed a 1.85-fold greater AUC and no detectable CNS toxicity at 100 mg/kg. In vitro hepatic metabolism studies showed that 22 was metabolized via cytochrome P-450, as previously shown, and by the flavin-containing monooxygenase (FMO). Deuterated-22 showed decreased metabolism and showed acceptable cardiovascular and physicochemical properties.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Mexiletine/análogos & derivados , Mexiletine/farmacocinética , Miocitos Cardíacos/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Células Cultivadas , Femenino , Humanos , Hígado/metabolismo , Síndrome de QT Prolongado , Masculino , Mexiletine/efectos adversos , Ratones Endogámicos BALB C , Ratas Sprague-Dawley , Convulsiones/inducido químicamente
19.
J Med Chem ; 64(9): 5384-5403, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33942619

RESUMEN

Ventricular cardiac arrhythmia (VA) arises in acquired or congenital heart disease. Long QT syndrome type-3 (LQT3) is a congenital form of VA caused by cardiac sodium channel (INaL) SCN5A mutations that prolongs cardiac action potential (AP) and enhances INaL current. Mexiletine inhibits INaL and shortens the QT interval in LQT3 patients. Above therapeutic doses, mexiletine prolongs the cardiac AP. We explored structure-activity relationships (SAR) for AP shortening and prolongation using dynamic medicinal chemistry and AP kinetics in human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Using patient-derived LQT3 and healthy hiPSC-CMs, we resolved distinct SAR for AP shortening and prolongation effects in mexiletine analogues and synthesized new analogues with enhanced potency and selectivity for INaL. This resulted in compounds with decreased AP prolongation effects, increased metabolic stability, increased INaL selectivity, and decreased avidity for the potassium channel. This study highlights using hiPSC-CMs to guide medicinal chemistry and "drug development in a dish".


Asunto(s)
Antiarrítmicos/química , Trastorno del Sistema de Conducción Cardíaco/patología , Síndrome de QT Prolongado/patología , Mexiletine/análogos & derivados , Potenciales de Acción/efectos de los fármacos , Animales , Antiarrítmicos/farmacología , Conducta Animal/efectos de los fármacos , Trastorno del Sistema de Conducción Cardíaco/metabolismo , Células Cultivadas , Diseño de Fármacos , Estabilidad de Medicamentos , Semivida , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Síndrome de QT Prolongado/metabolismo , Masculino , Mexiletine/farmacología , Ratones , Ratones Endogámicos BALB C , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad
20.
Cell Chem Biol ; 28(5): 625-635.e5, 2021 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-33503403

RESUMEN

Wnt signaling plays a central role in tissue maintenance and cancer. Wnt activates downstream genes through ß-catenin, which interacts with TCF/LEF transcription factors. A major question is how this signaling is coordinated relative to tissue organization and renewal. We used a recently described class of small molecules that binds tubulin to reveal a molecular cascade linking stress signaling through ATM, HIPK2, and p53 to the regulation of TCF/LEF transcriptional activity. These data suggest a mechanism by which mitotic and genotoxic stress can indirectly modulate Wnt responsiveness to exert coherent control over cell shape and renewal. These findings have implications for understanding tissue morphogenesis and small-molecule anticancer therapeutics.


Asunto(s)
Sondas Moleculares/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Factores de Transcripción TCF/antagonistas & inhibidores , beta Catenina/antagonistas & inhibidores , Animales , Células Cultivadas , Humanos , Masculino , Sondas Moleculares/química , Bibliotecas de Moléculas Pequeñas/química , Factores de Transcripción TCF/genética , Factores de Transcripción TCF/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Xenopus , Pez Cebra , beta Catenina/genética , beta Catenina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA