Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Adv Nutr ; 9(4): 524S-532S, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30032231

RESUMEN

The ability of certain foods to impair or augment the absorption of various vitamins and minerals has been recognized for many years. However, the contribution of botanical dietary supplements (BDSs) to altered micronutrient disposition has received little attention. Almost half of the US population uses some type of dietary supplement on a regular basis, with vitamin and mineral supplements constituting the majority of these products. BDS usage has also risen considerably over the last 2 decades, and a number of clinically relevant herb-drug interactions have been identified during this time. BDSs are formulated as concentrated plant extracts containing a plethora of unique phytochemicals not commonly found in the normal diet. Many of these uncommon phytochemicals can modulate various xenobiotic enzymes and transporters present in both the intestine and liver. Therefore, it is likely that the mechanisms underlying many herb-drug interactions can also affect micronutrient absorption, distribution, metabolism, and excretion. To date, very few prospective studies have attempted to characterize the prevalence and clinical relevance of herb-micronutrient interactions. Current research indicates that certain BDSs can reduce iron, folate, and ascorbate absorption, and others contribute to heavy metal intoxication. Researchers in the field of nutrition may not appreciate many of the idiosyncrasies of BDSs regarding product quality and dosage form performance. Failure to account for these eccentricities can adversely affect the outcome and interpretation of any prospective herb-micronutrient interaction study. This review highlights several clinically relevant herb-micronutrient interactions and describes several common pitfalls that often beset clinical research with BDSs.


Asunto(s)
Interacciones Farmacológicas , Micronutrientes/administración & dosificación , Minerales/administración & dosificación , Preparaciones de Plantas/administración & dosificación , Vitaminas/administración & dosificación , Suplementos Dietéticos , Humanos , Absorción Intestinal/efectos de los fármacos , Metales/administración & dosificación , Metales/toxicidad , Micronutrientes/farmacocinética , Minerales/farmacocinética , Fitoquímicos/administración & dosificación , Extractos Vegetales/administración & dosificación , Estudios Prospectivos , Estados Unidos , Vitaminas/farmacocinética
2.
Acta Crystallogr E Crystallogr Commun ; 73(Pt 6): 864-866, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28638646

RESUMEN

The title compound, C28H32N2, (I), is one of a second generation of compounds designed and synthesized based on a very potent and selective α9α10 nicotinic acetyl-choline receptor antagonist ZZ161C {1,1'-[[1,1'-biphen-yl]-4,4'-diylbis(prop-2-yne-3,1-di-yl)]bis-(3,4-di-methyl-pyridin-1-ium) bromide}, which has shown analgesic effects in a chemotherapy-induced neuropathy animal model. Compound (I) was synthesized by the reaction of 4,4'-bis-(3-bromo-prop-1-yn-1-yl)-1,1'-biphenyl with piperidine at room temperature in aceto-nitrile. The single-crystal used for X-ray analysis was obtained by dissolving (I) in a mixture of di-chloro-methane and methanol, followed by slow evaporation of the solvent. In the crystal of (I), the biphenyl moiety has a twisted conformation, with a dihedral angle of 25.93 (4)° between the benzene rings. Both piperidine head groups in (I) are in the chair conformation and are oriented so that the N-atom lone pairs of each piperidine group point away from the central biphenyl moiety.

3.
Acta Crystallogr E Crystallogr Commun ; 71(Pt 10): 1132-5, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26594389

RESUMEN

As part of a comprehensive program to discover α9α10 nicotinic acetyl-choline receptor antagonists, the title compounds C30H36N2, (I), and C36H48N2, (II), were synthesized by coupling 4,4'-bis-(3-bromo-prop-1-yn-1-yl)-1,1'-biphenyl with 4-methyl-piperidine and 2,2,6,6-tetra-methyl-piperidine, respectively, in aceto-nitrile at room temperature. In compound (I), the biphenyl system has a twisted conformation with a dihedral angle of 26.57 (6)° between the two phenyl rings of the biphenyl moiety, while in compound (II), the biphenyl moiety sits on a crystallographic inversion centre so the two phenyl rings are exactly coplanar. The terminal piperidine rings in both compound (I) and compound (II) are in the chair conformation. In compound (I), the dihedral angles about the ethynyl groups between the planes of the phenyl rings and the piperidine ring N atoms are 37.16 (16) and 14.20 (17)°. In compound (II), the corresponding dihedral angles are both 61.48 (17)°. There are no noteworthy inter-molecular inter-actions in (I), but in (II) there is a small π-overlap between inversion-related mol-ecules (1 - x, 1 - y, 1 - z), with an inter-planar spacing of 3.553 (3) Šand centroid-to-centroid separation of 3.859 (4) Å.

4.
Acta Crystallogr E Crystallogr Commun ; 71(Pt 10): 1147-50, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26594393

RESUMEN

The title compounds, C26H28N2, (I), and C28H32N2, (II), were designed based on the structure of the potent α9α10 nicotinic acetyl-choline receptor antagonist ZZ161C {1,1'-[[1,1'-biphen-yl]-4,4'-diylbis(prop-2-yne-3,1-di-yl)]bis-(3,4-di-methyl-pyridin-1-ium) bromide}. In order to improve the druglikeness properties of ZZ161C for potential oral administration, the title compounds (I) and (II) were prepared by coupling 4,4'-bis-(3-bromo-prop-1-yn-1-yl)-1,1'-biphenyl with pyrrol-idine, (I), and (S)-2-methyl-pyrrolidine, (II), respectively, in aceto-nitrile at room temperature. The asymmetric unit of (I) contains two half mol-ecules that each sit on sites of crystallographic inversion. As a result, the biphenyl ring systems in compound (I) are coplanar. The biphenyl ring system in compound (II), however, has a dihedral angle of 28.76 (11)°. In (I), the two independent mol-ecules differ in the orientation of the pyrrolidine ring (the nitro-gen lone pair points towards the biphenyl rings in one mol-ecule, but away from the rings in the other). The torsion angles about the ethynyl groups between the planes of the phenyl rings and the pyrrolidine ring N atoms are 84.15 (10) and -152.89 (10)°. In compound (II), the corresponding torsion angles are 122.0 (3) and 167.0 (3)°, with the nitro-gen lone pairs at both ends of the mol-ecule directed away from the central biphenyl rings.

5.
Biochem Pharmacol ; 97(3): 341-9, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26225832

RESUMEN

Risk assessment for exposure to mixtures of drugs and pollutants relies heavily on in vitro characterization of their bioactivation and/or metabolism individually and extrapolation to mixtures assuming no interaction. Herein, we demonstrated that in vitro CYP2E1 metabolic activation of acetaminophen and styrene mixtures could not be explained through the Michaelis-Menten mechanism or any models relying on that premise. As a baseline for mixture studies with styrene, steady-state analysis of acetaminophen oxidation revealed a biphasic kinetic profile that was best described by negative cooperativity (Hill coefficient=0.72). The best-fit mechanism for this relationship involved two binding sites with differing affinities (Ks=830µM and Kss=32mM). Introduction of styrene inhibited that reaction less than predicted by simple competition and thus provided evidence for a cooperative mechanism within the mixture. Likewise, acetaminophen acted through a mixed-type inhibition mechanism to impact styrene epoxidation. In this case, acetaminophen competed with styrene for CYP2E1 (Ki=830µM and Ksi=180µM for catalytic and effector sites, respectively) and resulted in cooperative impacts on binding and catalysis. Based on modeling of in vivo clearance, cooperative interactions between acetaminophen and styrene resulted in profoundly increased styrene activation at low styrene exposure levels and therapeutic acetaminophen levels. Current Michaelis-Menten based toxicological models for mixtures such as styrene and acetaminophen would fail to detect this concentration-dependent relationship. Hence, future studies must assess the role of alternate CYP2E1 mechanisms in bioactivation of compounds to improve the accuracy of interpretations and predictions of toxicity.


Asunto(s)
Acetaminofén/metabolismo , Inhibidores del Citocromo P-450 CYP2E1/metabolismo , Citocromo P-450 CYP2E1/metabolismo , Contaminantes Ambientales/metabolismo , Microsomas Hepáticos/enzimología , Estireno/metabolismo , Acetaminofén/química , Acetaminofén/toxicidad , Sitios de Unión , Unión Competitiva , Biotransformación , Inhibidores del Citocromo P-450 CYP2E1/química , Inhibidores del Citocromo P-450 CYP2E1/toxicidad , Contaminantes Ambientales/química , Contaminantes Ambientales/toxicidad , Humanos , Técnicas In Vitro , Cinética , Microsomas Hepáticos/efectos de los fármacos , Modelos Biológicos , Modelos Químicos , Oxidación-Reducción , Estireno/química , Estireno/toxicidad , Especificidad por Sustrato
6.
Drug Metab Dispos ; 43(4): 534-52, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25609220

RESUMEN

The disposition and metabolism of hydrastine was investigated in 11 healthy subjects following an oral dose of 2.7 g of goldenseal supplement containing 78 mg of hydrastine. Serial blood samples were collected for 48 hours, and urine was collected for 24 hours. Hydrastine serum and urine concentrations were determined by Liquid Chromatography-tandem mass spectrometry (LC-MS/MS). Pharmacokinetic parameters for hydrastine were calculated using noncompartmental methods. The maximal serum concentration (Cmax) was 225 ± 100 ng/ml, Tmax was 1.5 ± 0.3 hours, and area under the curve was 6.4 ± 4.1 ng ⋅ h/ml ⋅ kg. The elimination half-life was 4.8 ± 1.4 hours. Metabolites of hydrastine were identified in serum and urine by using liquid chromatography coupled to high-resolution mass spectrometry. Hydrastine metabolites were identified by various mass spectrometric techniques, such as accurate mass measurement, neutral loss scanning, and product ion scanning using Quadrupole-Time of Flight (Q-ToF) and triple quadrupole instruments. The identity of phase II metabolites was further confirmed by hydrolysis of glucuronide and sulfate conjugates using bovine ß-glucuronidase and a Helix pomatia sulfatase/glucuronidase enzyme preparation. Hydrastine was found to undergo rapid and extensive phase I and phase II metabolism. Reduction, O-demethylation, N-demethylation, hydroxylation, aromatization, lactone hydrolysis, and dehydrogenation of the alcohol group formed by lactone hydrolysis to the ketone group were observed during phase I biotransformation of hydrastine. Phase II metabolites were primarily glucuronide and sulfate conjugates. Hydrastine undergoes extensive biotransformation, and some metabolites may have pharmacological activity. Further study is needed in this area.


Asunto(s)
Bencilisoquinolinas/sangre , Bencilisoquinolinas/orina , Suplementos Dietéticos , Hydrastis/química , Administración Oral , Bencilisoquinolinas/administración & dosificación , Bencilisoquinolinas/metabolismo , Cromatografía Liquida , Estabilidad de Medicamentos , Femenino , Voluntarios Sanos , Humanos , Masculino , Fase I de la Desintoxicación Metabólica , Fase II de la Desintoxicación Metabólica , Proyectos Piloto , Espectrometría de Masas en Tándem , Distribución Tisular
7.
Mol Pharmacol ; 84(3): 459-75, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23839567

RESUMEN

Positive allosteric modulators (PAMs) of α7 nicotinic acetylcholine receptors can enhance ion channel currents and downstream effects of α7 stimulation. We investigated the approach of using noncompetitive antagonists to regulate α7 receptor function, potentially distinguishing effects requiring ion channel currents from signaling induced by nonconducting states. Three small readily reversible antagonists, (1S,2R,4R)-N,2,3,3-tetramethylbicyclo[2.2.1]heptan-2-amine (mecamylamine), N-(2.6-dimethylphenylcarbamoylmethyl)triethylammonium bromide (QX-314), and 2-(dimethylamino)ethyl 4-(butylamino)benzoate (tetracaine), as well as three large slowly reversible antagonists, bis-(2,2,6,6-tetramethyl-4-piperidinyl) sebacate (BTMPS), 2,2,6,6-tetramethylpiperidin-4-yl heptanoate (TMPH), and 1,2,4,5-tetra-{5-[1-(3-benzyl)pyridinium]pent-1-yl}benzene tetrabromide (tkP3BzPB), were investigated for their effectiveness and voltage dependence in the inhibition of responses evoked by acetylcholine alone or augmented by the α7-selective PAM N-(5-chloro-2,4-dimethoxyphenyl)-N'-(5-methyl-3-isoxazolyl)-urea (PNU-120596). Analyses of the small antagonists on PNU-120596-potentiated single-channel bursts indicated that each agent had a distinct mechanism of inhibition and only that of QX-314 was consistent with simple open channel block. In addition to decreasing channel open times and burst durations, mecamylamine and tetracaine induced unique subconductance states. To determine whether channel-blocking activity alone would be sufficient to prevent cell death, the antagonists were tested for their ability to protect α7-expressing cells from cytotoxic effects of the α7 agonist choline in combination with PNU-120596. Only tetracaine and tkP3BzPB, the two agents that had effects least consistent with simple ion channel block, were fully cytoprotective at concentrations that gave submaximal inhibition of macroscopic currents in oocytes. Further analyses indicated that toxicity produced by PNU-120596 and choline was calcium independent and likely an apoptotic event. Our results are consistent with the hypothesis that PAMs may modulate conformational states important for both channel activity and ion channel-independent signaling.


Asunto(s)
Agonistas Nicotínicos/farmacología , Antagonistas Nicotínicos/farmacología , Receptor Nicotínico de Acetilcolina alfa 7/fisiología , Regulación Alostérica , Animales , Calcio/metabolismo , Línea Celular , Colina/farmacología , Citotoxinas/farmacología , Femenino , Humanos , Isoxazoles/farmacología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Oocitos/efectos de los fármacos , Oocitos/fisiología , Técnicas de Placa-Clamp , Compuestos de Fenilurea/farmacología , Xenopus laevis
8.
Anal Chem ; 83(16): 6381-8, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21740038

RESUMEN

The aminoalkylindole agonists JWH-018 and JWH-073 are contained in "K2/SPICE" products sold as "legal marijuana". Previous human metabolic studies have identified (ω)-hydroxyl and (ω)-carboxyl metabolites as biomarkers that are indicative of product use. However, other primary metabolites exhibiting similar chromatographic properties and mass spectra are also excreted in human urine. Analytical standards were used in this study to identify new primary metabolites as (ω-1)-hydroxyl derivatives of JWH-018 and JWH-073. The liquid chromatography tandem mass spectrometry (LC-MS/MS) procedure, coupled with an automated solid-phase extraction procedure incorporating deuterium-labeled internal standards, provides rapid resolution of the (ω)- and (ω-1) metabolites with adequate sensitivity, precision, and accuracy for trace analysis in human urine. Results from four urine specimens collected after individuals reportedly self-administered either JWH-018 or a mixture of JWH-018 and JWH-073 showed the following: (1) all tested metabolites were excreted in high concentrations, (2) (ω)- and (ω-1)-hydroxyl metabolites were exclusively excreted as glucuronic acid conjugates, and (3) ∼5%-80% of the (ω)-carboxyl metabolites was excreted as glucuronic acid conjugates. This is the first report to identify and quantify (ω-1)-hydroxyl metabolites of JWH-018 and JWH-073 and the first to incorporate automated extraction procedures using deuterium-labeled internal standards. Full clinical validation awaits further testing.


Asunto(s)
Cromatografía Liquida/métodos , Drogas Ilícitas/orina , Indoles/orina , Naftalenos/orina , Extracción en Fase Sólida/métodos , Detección de Abuso de Sustancias/métodos , Espectrometría de Masas en Tándem/métodos , Deuterio/química , Humanos , Estándares de Referencia , Sensibilidad y Especificidad , Coloración y Etiquetado
9.
J Pharmacol Exp Ther ; 337(1): 110-6, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21205919

RESUMEN

In overdose the analgesic/antipyretic acetaminophen (APAP) is hepatotoxic. Toxicity is mediated by initial hepatic metabolism to N-acetyl-p-benzoquinone imine (NAPQI). After low doses NAPQI is efficiently detoxified by GSH. However, in overdose GSH is depleted, NAPQI covalently binds to proteins as APAP adducts, and oxygen/nitrogen stress occurs. Toxicity is believed to occur by mitochondrial dysfunction. Manganese superoxide dismutase (MnSOD) inactivation by protein nitration has been reported to occur during other oxidant stress-mediated diseases. MnSOD is a critical mitochondrial antioxidant enzyme that prevents peroxynitrite formation within the mitochondria. To examine the role of MnSOD in APAP toxicity, mice were treated with 300 mg/kg APAP. GSH was significantly reduced by 65% at 0.5 h and remained reduced from 1 to 4 h. Serum alanine aminotransferase did not significantly increase until 4 h and was 2290 IU/liter at 6 h. MnSOD activity was significantly reduced by 50% at 1 and 2 h. At 1 h, GSH was significantly depleted by 62 and 80% at nontoxic doses of 50 and 100 mg/kg, respectively. No further GSH depletion occurred with hepatotoxic doses of 200 and 300 mg/kg APAP. A dose response decrease in MnSOD activity was observed for APAP at 100, 200, and 300 mg/kg. Immunoprecipitation of MnSOD from livers of APAP-treated mice followed by Western blot analysis revealed nitrated MnSOD. APAP-MnSOD adducts were not detected. Treatment of recombinant MnSOD with NAPQI did not produce APAP protein adducts. The data indicate that MnSOD inactivation by nitration is an early event in APAP-induced hepatic toxicity.


Asunto(s)
Acetaminofén/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/enzimología , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/enzimología , Superóxido Dismutasa/antagonistas & inhibidores , Superóxido Dismutasa/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Nitratos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología
10.
Drug Dev Ind Pharm ; 35(1): 1-11, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18991061

RESUMEN

A water-insoluble complex between diltiazem HCl and Na deoxycholate was prepared to achieve sustained release dosage forms. Physicochemical characterization of the drug complex was carried out with differential scanning calorimetry, (1)H-nuclear magnetic resonance, and Fourier transform infrared spectroscopy. These techniques showed that the characteristic peaks in both the drug and the complexing agent (protonated amine and carboxylate) disappeared and new peaks appeared upon formation of the ionic complex. The release of diltiazem from drug-complex tablets was sustained for a long period of time (>24 h) and was dependent on the pH of the dissolution medium. However, the dependence of drug release on pH was eliminated at pH 6-8 and minimized at pH 1.5 when drug-complex powders were incorporated in hydroxypropylmethylcellulose (HPMC) drug carriers. Unlike the release of diltiazem HCl from HPMC drug carriers, drug release from drug-complex/HPMC tablets was linear or near linear irrespective of the viscosity grade of the polymer (E15 to K4M). This is due to a shift in the controlling mechanism of drug release from drug diffusion to erosion of polymer. Also, drug release kinetics was not significantly affected by the water solubility of cationic drugs (diltiazem HCl, verapamil HCl, propranolol HCl, and labetalol HCl) ranging from 1.6 to 62% and the type of amine (i.e., secondary or tertiary). The same release characteristics were observed from the complexes between anionic drugs (Na salicylate, naproxen Na, and tolmetin Na) and benzathine diacetate as found from the complexes between cationic drugs and Na deoxycholate.


Asunto(s)
Ácido Desoxicólico/química , Diltiazem/química , Portadores de Fármacos/química , Electrólitos/química , Rastreo Diferencial de Calorimetría , Química Farmacéutica , Preparaciones de Acción Retardada , Etilenodiaminas/química , Concentración de Iones de Hidrógeno , Derivados de la Hipromelosa , Espectroscopía de Resonancia Magnética , Metilcelulosa/análogos & derivados , Metilcelulosa/química , Solubilidad , Espectroscopía Infrarroja por Transformada de Fourier , Comprimidos , Factores de Tiempo , Viscosidad
11.
Am J Physiol Renal Physiol ; 294(2): F423-32, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18077599

RESUMEN

Previously, we showed that protein kinase B (Akt) activation increases intracellular ATP levels and decreases necrosis in renal proximal tubular cells (RPTC) injured by the nephrotoxicant S-(1, 2-dichlorovinyl)-l-cysteine (DCVC) (Shaik ZP, Fifer EK, Nowak G. Am J Physiol Renal Physiol 292: F292-F303, 2007). This study examined the role of Akt in improving mitochondrial function in DCVC-injured RPTC. Our data show a novel observation that phosphorylated (active) Akt is localized in mitochondria of noninjured RPTC, both in mitoplasts and the mitochondrial outer membrane. Mitochondrial levels of active Akt decreased in nephrotoxicant-injured RPTC, and this decrease was associated with mitochondrial dysfunction. DCVC decreased basal, uncoupled, and state 3 respirations; ATP production; activities of complexes I, II, and III; the mitochondrial membrane potential (DeltaPsi(m)); and F(0)F(1)-ATPase activity. Expressing constitutively active Akt in DCVC-injured RPTC increased the levels of phosphorylated Akt in mitochondria, reduced the decreases in basal and uncoupled respirations, increased complex I-coupled state 3 respiration and ATP production, enhanced activities of complex I, complex III, and F(0)F(1)-ATPase, and improved DeltaPsi(m). In contrast, inhibiting Akt activation by expressing dominant negative (inactive) Akt or using 20 microM LY294002 exacerbated decreases in electron transport rate, state 3 respiration, ATP production, DeltaPsi(m), and activities of complex I, complex III, and F(0)F(1)-ATPase. In conclusion, our data show that Akt activation promotes mitochondrial respiration and ATP production in toxicant-injured RPTC by 1) improving integrity of the respiratory chain and maintaining activities of complex I and complex III, 2) reducing decreases in DeltaPsi(m), and 3) restoring F(0)F(1)-ATPase activity.


Asunto(s)
Enfermedades Renales/metabolismo , Túbulos Renales Proximales/citología , Mitocondrias/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Adenosina Trifosfato/biosíntesis , Animales , Células Cultivadas , Cromonas/farmacología , Cisteína/análogos & derivados , Cisteína/toxicidad , Transporte de Electrón/efectos de los fármacos , Transporte de Electrón/fisiología , Complejo I de Transporte de Electrón/efectos de los fármacos , Complejo I de Transporte de Electrón/metabolismo , Complejo II de Transporte de Electrones/efectos de los fármacos , Complejo II de Transporte de Electrones/metabolismo , Complejo III de Transporte de Electrones/efectos de los fármacos , Complejo III de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/efectos de los fármacos , Complejo IV de Transporte de Electrones/metabolismo , Contaminantes Ambientales/toxicidad , Inhibidores Enzimáticos/farmacología , Enfermedades Renales/inducido químicamente , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/fisiopatología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/fisiología , Mitocondrias/efectos de los fármacos , Membranas Mitocondriales/metabolismo , Morfolinas/farmacología , Oligomicinas/farmacología , Fosforilación Oxidativa/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Fosforilación/efectos de los fármacos , ATPasas de Translocación de Protón/antagonistas & inhibidores , ATPasas de Translocación de Protón/efectos de los fármacos , ATPasas de Translocación de Protón/metabolismo , Conejos , Transfección
12.
Am J Physiol Renal Physiol ; 292(1): F292-303, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16940564

RESUMEN

Protein kinase B (Akt) activation is well known for its protective effects against apoptosis. However, the role of Akt in regulation of necrosis is unknown. This study was designed to test whether Akt activation protects against nephrotoxicant-induced injury and death in renal proximal tubular cells (RPTC). Exposure of primary cultures of RPTC to the nephrotoxic cysteine conjugate, S-(1,2-dichlorovinyl)-l-cysteine (DCVC), resulted in 9% apoptosis and 30% necrosis at 24 h following the exposure. Akt was activated during 8 h but not at 24 h following toxicant exposure. No RPTC necrosis was observed during Akt activation. Blocking Akt activation using a phosphatidylinositol 3-kinase inhibitor, LY294002 (20 muM), or expressing dominant negative (inactive) Akt increased DCVC-induced RPTC necrosis to 42%. In contrast, Akt activation by expression of constitutively active Akt diminished necrosis to 15%. Modulation of Akt activity had no effect on DCVC-induced apoptosis. DCVC-induced RPTC injury was accompanied by decreases in respiration (51% of controls) and ATP levels (57% of controls). Akt inhibition exacerbated decreases in RPTC respiration and intracellular ATP content (both to 30% of controls). In contrast, Akt activation reduced DCVC-induced decreases in respiration (80% of controls) and prevented decline in ATP content. These data show that in RPTC, Akt activation reduces 1) toxicant-induced mitochondrial dysfunction, 2) decreases in ATP levels, and 3) necrosis. We conclude that Akt activation plays a protective role against necrosis caused by nephrotoxic insult in RPTC. Furthermore, we identified mitochondria as a subcellular target of protective actions of Akt against necrosis.


Asunto(s)
Enfermedades Renales/inducido químicamente , Enfermedades Renales/patología , Proteína Oncogénica v-akt/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Adenosina Trifosfato/metabolismo , Adenoviridae/genética , Animales , Muerte Celular , Células Cultivadas , Cisteína/análogos & derivados , Cisteína/toxicidad , ADN Complementario/biosíntesis , ADN Complementario/genética , Femenino , Colorantes Fluorescentes , Immunoblotting , Inmunohistoquímica , Indoles , Túbulos Renales/patología , L-Lactato Deshidrogenasa/metabolismo , Mitocondrias/metabolismo , Necrosis , Proteína Oncogénica v-akt/biosíntesis , Proteína Oncogénica v-akt/genética , Consumo de Oxígeno/efectos de los fármacos , Conejos , Transfección
13.
Life Sci ; 78(19): 2159-65, 2006 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-16318858

RESUMEN

This study was designed to examine the potential structure-activity relationship between carboxylic acids, histone acetylation and T cell cycle blockade. Toward this goal a series of structural homologues of the short-chain carboxylic acid n-butyrate were studied for their ability to block the IL-2-stimulated proliferation of cloned CD4+ T cells. The carboxylic acids were also tested for their ability to inhibit histone deacetylation. In addition, Western blotting was used to examine the relative capacity of the carboxlic acids to upregulate the cyclin kinase-dependent inhibitor p21cip1 in T cells. As shown earlier n-butyrate effectively inhibited histone deacetylation. The increased acetylation induced by n-butyrate was associated with the upregulation of the cyclin-dependent kinase inhibitor p21cip1 and the cell cycle blockade of CD4+ T cells. Of the other carboxylic acids studied, the short chain acids, C3-C5, without branching were the best inhibitors of histone deacetylase. This inhibition correlated with increased expression of the cell cycle blocker p21cip1, and the associated suppression of CD4+ T cell proliferation. The branched-chain carboxylic acids tested were ineffective in all the assays. These results underline the relationship between the ability of a carboxylic acid to inhibit histone deacetylation, and their ability to block T cell proliferation, and suggests that branching inhibits these effects.


Asunto(s)
Ácidos Carboxílicos/farmacocinética , Proliferación Celular/efectos de los fármacos , Células TH1/efectos de los fármacos , Acetilación , Animales , Ácidos Carboxílicos/química , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Histonas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Relación Estructura-Actividad , Células TH1/citología , Regulación hacia Arriba
14.
J Cancer Educ ; 21(4): 243-7, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17542717

RESUMEN

BACKGROUND: The Partners in Research program provides first-hand research experiences for medical, pharmacy, and African-American undergraduate students. METHODS: During ten weeks, students participate in on-going cancer research, weekly educational sessions, two observational clinic sessions, and at least one patient support-community outreach clinic. RESULTS: Over the past six years the program has enrolled 155 students. Surveys indicate that most students give the course high ratings and would recommend the course to their peers. Follow-up shows their continued interest in research. CONCLUSIONS: The program will encourage students to pursue careers in cancer research and provide a solid base of knowledge to future health care professionals.


Asunto(s)
Investigación Biomédica/organización & administración , Educación de Pregrado en Medicina/organización & administración , Oncología Médica/educación , Selección de Profesión , Humanos
15.
Immunopharmacol Immunotoxicol ; 27(4): 545-64, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16435576

RESUMEN

MEB [n-butyrate 2-(4-morpholinyl) ethyl butyrate hydrochloride], a histone deacetylase inhibitor and G1 blocker, has been shown to induce unresponsiveness in antigen-activated Th1 cells. MEB was tested for here for its ability to inactivate naive alloantigen-specific T cells from DBA/2 and C57BL/10 mice. Since T cells from these two strains of mice have been shown to differ in their cell cycle regulation, it we hoped that this comparison would provide information concerning the role of cycle regulatory proteins in mediating MEB-induced T cell unresponsiveness. MEB inhibited proliferation in a one-way mixed lymphocyte reaction (MLR) in which spleen cells from DBA/2 mice (H-2d) or C57BL/10 mice (H-2b) were stimulated with spleen cells from C57BL/10 or DBA/2 mice, respectively. C57BL/10 responder T cells isolated from the MEB-treated primary MLR remained unresponsive to alloantigen following restimulation in a secondary MLR that did not contain MEB. T cells from DBA/2 mice were less sensitive to MEB-induced unresponsiveness and required a longer exposure or pretreatment with IL-2 to become tolerant. In all cases responsiveness to MEB-induced tolerance in the alloantigen-stimulated T cells corresponded with the levels of the cyclin-dependent kinase inhibitor p21cip1. Additional experiments showed that T cells from p21cip1-deficient mice, unlike T cells from p21cip1 wild-type littermates, were resistant to MEB-induced tolerance. These results underscore the role of p21cip1 in mediating T cell tolerance induced by the histone deacetylase inhibitor MEB.


Asunto(s)
Butiratos/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidores de Histona Desacetilasas , Morfolinas/farmacología , Subgrupos de Linfocitos T/efectos de los fármacos , Animales , Linfocitos T CD4-Positivos/enzimología , Linfocitos T CD8-positivos/enzimología , Ciclo Celular/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Relación Dosis-Respuesta a Droga , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Células Jurkat , Prueba de Cultivo Mixto de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Especificidad de la Especie , Bazo/citología , Bazo/efectos de los fármacos , Bazo/enzimología , Subgrupos de Linfocitos T/enzimología , Factores de Tiempo
16.
Int Immunopharmacol ; 4(9): 1231-9, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15251119

RESUMEN

The butyric acid derivative, 2-(4-morpholynl) ethyl butyrate hydrochloride (MEB), has been reported to induce antigen-specific T cell unresponsiveness and to block T cell-mediated graft-versus-host disease. As a potential therapeutic agent, it was important to determine the effects of MEB on other cells that contribute to immunopathology. Accordingly, we tested the effects of MEB on macrophage functions. MEB did not affect macrophage viability, phagocytic activity, or the activation-induced up-regulation of molecules associated with antigen presentation: MHC-II, CD86, CD40, or ICAM-1. However, MEB potently inhibited activation-induced production of inflammatory mediators, including tumor necrosis factor-alpha (TNF-alpha), IL-6, chemokine CCL2 and nitric oxide (NO). MEB inhibited the induction of NO synthase (NOS2), which is necessary for inducible NO, and inhibited nuclear translocation of NFkappaB, suggesting that MEB interferes with the signaling pathway involved in NOS2 induction. Thus, while inducing specific T cell unresponsiveness, MEB also exerts anti-inflammatory activity by acting on macrophages to suppress production of cytokines and NO.


Asunto(s)
Antígenos/inmunología , Butiratos/farmacología , Antagonistas de los Receptores Histamínicos/farmacología , Mediadores de Inflamación/metabolismo , Macrófagos/metabolismo , Morfolinas/farmacología , Linfocitos T/efectos de los fármacos , Animales , Antígenos CD/biosíntesis , Antígeno B7-2 , Western Blotting , Antígenos CD40/biosíntesis , Células Cultivadas , Citotoxicidad Inmunológica/efectos de los fármacos , Femenino , Citometría de Flujo , Genes MHC Clase II/efectos de los fármacos , Molécula 1 de Adhesión Intercelular/biosíntesis , Activación de Linfocitos/inmunología , Activación de Linfocitos/fisiología , Macrófagos/efectos de los fármacos , Glicoproteínas de Membrana/biosíntesis , Ratones , Ratones Endogámicos C57BL , FN-kappa B/efectos de los fármacos , Óxido Nítrico/metabolismo , Fagocitosis/efectos de los fármacos
17.
Immunopharmacol Immunotoxicol ; 25(1): 13-27, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12675196

RESUMEN

Graft-versus-host-disease (GVHD) is a common and potentially fatal complication following bone marrow transplantation. This study was initiated to test whether MEB [n-butyrate 2-(4-morpholinyl) ethyl butyrate hydrochloride], a derivative of the G1 blocker butyric acid, could specifically inactivate the alloantigen-specific T cells that mediate GVHD. MEB was shown to inhibit proliferation in a one-way mixed lymphocyte reaction (MLR) in which spleen cells from C57BL/6 mice (H-2b) were stimulated with spleen cells from DBA/2 mice (H-2d). The addition of MEB to the MLR prevented the expansion of alloantigen-stimulated CD8+ and CD4+ T cells in association with decreased IL-2 production. In addition, MEB inhibited the CTL activity of CD8+ T cells from the MLR. Most importantly, T cells from the MEB-treated MLR, unlike T cells from an untreated MLR, were unable to induce the splenomegaly and increased serum TNF-alpha levels characteristic of acute GVHD when injected into B6D2F1 mice. The splenomegaly found in the B6D2F1 mice injected with T cells from an untreated MLR encompassed the expansion and activation of CD8+ T cells, CD4+ T cells, B cells and macrophages. In contrast, the spleens of mice injected with T cells from MEB-treated MLR looked essentially identical to those of control B6D2F1 mice in terms of the numbers and activation state of the spleen cell populations. Similarly, the increase in IFN-gamma and TNF-alpha production by CD4+ and CD8+ T cells from the spleens of mice undergoing acute GVHD was not observed if the mice were injected with T cells from an MEB-treated MLR instead of an untreated MLR. The use of MEB to inactivate host-specific T cells ex vivo underlines the potential clinical importance of this compound in the prevention and treatment of unwanted immune responses such as GVHD.


Asunto(s)
Butiratos/uso terapéutico , Anergia Clonal/efectos de los fármacos , Enfermedad Injerto contra Huésped/prevención & control , Isoantígenos/efectos de los fármacos , Morfolinas/uso terapéutico , Linfocitos T/efectos de los fármacos , Animales , Butiratos/farmacología , Antígenos CD4/efectos de los fármacos , Antígenos CD4/metabolismo , Antígenos CD8/efectos de los fármacos , Antígenos CD8/metabolismo , Trasplante de Células , Células Cultivadas , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Enfermedad Injerto contra Huésped/inmunología , Isoantígenos/inmunología , Activación de Linfocitos/efectos de los fármacos , Prueba de Cultivo Mixto de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Endogámicos , Morfolinas/farmacología , Bazo/citología , Bazo/efectos de los fármacos , Bazo/inmunología , Linfocitos T/inmunología , Factor de Necrosis Tumoral alfa/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA