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.
Anal Chem ; 94(2): 1158-1168, 2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-34958550

RESUMEN

Quantitative analysis of antibody-drug conjugates (ADCs) involves cleavage of ADCs into smaller analytes representing different components and subsequent measurements from multiple assays for a more comprehensive pharmacokinetic (PK) assessment. Multiple PK analytes including the drug remaining conjugated to the antibody (or antibody-conjugated drug, acDrug) and total antibody can be accessed simultaneously using a multiplex assay by proteolytic digestion of an ADC, if the sites of conjugation are homogeneous for an ADC and the linker drug is stable to proteases. Herein, a multiplexed immunoaffinity liquid chromatography-mass spectrometry (LC-MS)/MS PK assay is described involving immunoaffinity enrichment, enzymatic conversion of prodrug, trypsin digestion, and LC-MS/MS as applied to next-generation ADCs constructed from linker drugs bearing dimeric cyclopropabenzindole (CBI) payloads (duocarmycin analogues). The cytotoxic payload is chemically labile, requiring extensive optimization in sample preparation steps to stabilize the drug without ex vivo modification and to convert the prodrug into a single active form of the drug. The qualification data for this assay format showed that this approach provides robust acDrug and total antibody data and can be extended to ADCs with different monoclonal antibody frameworks and linker chemistries. Applications of this multiplexed assay to support preclinical studies are presented.


Asunto(s)
Antineoplásicos , Inmunoconjugados , Anticuerpos Monoclonales/química , Antineoplásicos/química , Cromatografía Liquida/métodos , Inmunoconjugados/química , Espectrometría de Masas en Tándem/métodos
2.
Mol Cancer Ther ; 20(2): 340-346, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33273056

RESUMEN

We are interested in developing a second generation of antibody-drug conjugates (ADCs) for the treatment of non-Hodgkin lymphoma (NHL) that could provide a longer duration of response and be more effective in indolent NHL than the microtubule-inhibiting ADCs pinatuzumab vedotin [anti-CD22-vc-monomethyl auristatin E (MMAE)] and polatuzumab vedotin (anti-CD79b-vc-MMAE). Pinatuzumab vedotin (anti-CD22-vc-MMAE) and polatuzumab vedotin (anti-CD79b-vc-MMAE) are ADCs that contain the microtubule inhibitor MMAE. Clinical trial data suggest that these ADCs have promising efficacy for the treatment of NHL; however, some patients do not respond or become resistant to the ADCs. We tested an anti-CD22 ADC with a seco-CBI-dimer payload, thio-Hu anti-CD22-(LC:K149C)-SN36248, and compared it with pinatuzumab vedotin for its efficacy and duration of response in xenograft models and its ability to deplete normal B cells in cynomolgus monkeys. We found that anti-CD22-(LC:K149C)-SN36248 was effective in xenograft models resistant to pinatuzumab vedotin, gave a longer duration of response, had a different mechanism of resistance, and was able to deplete normal B cells better than pinatuzumab vedotin. These studies provide evidence that anti-CD22-(LC:K149C)-SN36248 has the potential for longer duration of response and more efficacy in indolent NHL than MMAE ADCs and may provide the opportunity to improve outcomes for patients with NHL.


Asunto(s)
Aminobenzoatos/uso terapéutico , Inmunoconjugados/uso terapéutico , Linfoma no Hodgkin/tratamiento farmacológico , Oligopéptidos/uso terapéutico , Lectina 2 Similar a Ig de Unión al Ácido Siálico/metabolismo , Aminobenzoatos/farmacología , Animales , Línea Celular Tumoral , Haplorrinos , Humanos , Inmunoconjugados/farmacología , Oligopéptidos/farmacología
3.
Pharmacol Res Perspect ; 6(6): e00434, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30464842

RESUMEN

Although Interleukin-22 (IL-22) is produced by various leukocytes, it preferentially targets cells with epithelial origins. IL-22 exerts essential roles in modulating various tissue epithelial functions, such as innate host defense against extracellular pathogens, barrier integrity, regeneration, and wound healing. Therefore, IL-22 is thought to have therapeutic potential in treating diseases associated with infection, tissue injury or chronic tissue damage. A number of in vitro and in vivo nonclinical studies were conducted to characterize the pharmacological activity and safety parameters of UTTR1147A, an IL-22 recombinant fusion protein that links the human cytokine IL-22 with the Fc portion of a human immunoglobulin. To assess the pharmacological activity of UTTR1147A, STAT3 activation was evaluated in primary hepatocytes isolated from human, cynomolgus monkey, minipig, rat, and mouse after incubation with UTTR1147A. UTTR1147A activated STAT3 in all species evaluated, demonstrating that all were appropriate nonclinical species for toxicology studies. The nonclinical safety profile of UTTR1147A was evaluated in rats, minipigs, and cynomolgus monkeys to establish a safe clinical starting dose for humans in Phase I trials and to support clinical intravenous, subcutaneous and/or topical administration treatment regimen. Results demonstrate the cross-species translatability of the biological response in activating the IL-22 pathway as well as the translatability of findings from in vitro to in vivo systems. UTTR1147A was well tolerated in all species tested and induced the expected pharmacologic effects of epidermal hyperplasia and a transient increase in on-target acute phase proteins. These effects were all considered to be clinically predictable, manageable, monitorable, and reversible.


Asunto(s)
Hepatocitos/efectos de los fármacos , Factores Inmunológicos/farmacología , Interleucinas/toxicidad , Proteínas Recombinantes de Fusión/toxicidad , Animales , Ensayos Clínicos Fase I como Asunto , Evaluación Preclínica de Medicamentos , Femenino , Hepatocitos/metabolismo , Humanos , Interleucinas/administración & dosificación , Macaca fascicularis , Masculino , Ratones , Cultivo Primario de Células , Ratas , Proteínas Recombinantes de Fusión/administración & dosificación , Factor de Transcripción STAT3/metabolismo , Porcinos , Porcinos Enanos , Interleucina-22
4.
Sci Transl Med ; 7(273): 273ra15, 2015 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-25653221

RESUMEN

Inhibition of the kinase activity of leucine-rich repeat kinase 2 (LRRK2) is under investigation as a possible treatment for Parkinson's disease. However, there is no clinical validation as yet, and the safety implications of targeting LRRK2 kinase activity are not well understood. We evaluated the potential safety risks by comparing human and mouse LRRK2 mRNA tissue expression, by analyzing a Lrrk2 knockout mouse model, and by testing selective brain-penetrating LRRK2 kinase inhibitors in multiple species. LRRK2 mRNA tissue expression was comparable between species. Phenotypic analysis of Lrrk2 knockout mice revealed morphologic changes in lungs and kidneys, similar to those reported previously. However, in preclinical toxicity assessments in rodents, no pulmonary or renal changes were induced by two distinct LRRK2 kinase inhibitors. Both of these kinase inhibitors induced abnormal cytoplasmic accumulation of secretory lysosome-related organelles known as lamellar bodies in type II pneumocytes of the lung in nonhuman primates, but no lysosomal abnormality was observed in the kidney. The pulmonary change resembled the phenotype of Lrrk2 knockout mice, suggesting that this was LRRK2-mediated rather than a nonspecific or off-target effect. A biomarker of lysosomal dysregulation, di-docosahexaenoyl (22:6) bis(monoacylglycerol) phosphate (di-22:6-BMP), was also decreased in the urine of Lrrk2 knockout mice and nonhuman primates treated with LRRK2 kinase inhibitors. Our results suggest a role for LRRK2 in regulating lysosome-related lamellar bodies and that pulmonary toxicity may be a critical safety liability for LRRK2 kinase inhibitors in patients.


Asunto(s)
Pulmón/enzimología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/patología , Animales , Biomarcadores/sangre , Biomarcadores/orina , Relación Dosis-Respuesta a Droga , Femenino , Células HEK293 , Humanos , Riñón/anomalías , Riñón/efectos de los fármacos , Riñón/patología , Riñón/ultraestructura , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Pulmón/anomalías , Pulmón/patología , Pulmón/ultraestructura , Macaca fascicularis , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Morfolinas/química , Morfolinas/farmacología , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Pirazoles/química , Pirazoles/farmacología , Pirimidinas/química , Pirimidinas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley
5.
J Med Chem ; 57(3): 921-36, 2014 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-24354345

RESUMEN

Leucine-rich repeat kinase 2 (LRRK2) has drawn significant interest in the neuroscience research community because it is one of the most compelling targets for a potential disease-modifying Parkinson's disease therapy. Herein, we disclose structurally diverse small molecule inhibitors suitable for assessing the implications of sustained in vivo LRRK2 inhibition. Using previously reported aminopyrazole 2 as a lead molecule, we were able to engineer structural modifications in the solvent-exposed region of the ATP-binding site that significantly improve human hepatocyte stability, rat free brain exposure, and CYP inhibition and induction liabilities. Disciplined application of established optimal CNS design parameters culminated in the rapid identification of GNE-0877 (11) and GNE-9605 (20) as highly potent and selective LRRK2 inhibitors. The demonstrated metabolic stability, brain penetration across multiple species, and selectivity of these inhibitors support their use in preclinical efficacy and safety studies.


Asunto(s)
Encéfalo/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pirazoles/química , Pirimidinas/química , Animales , Línea Celular , Hepatocitos/metabolismo , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Macaca fascicularis , Microsomas Hepáticos/metabolismo , Simulación del Acoplamiento Molecular , Pirazoles/farmacocinética , Pirazoles/farmacología , Pirimidinas/farmacocinética , Pirimidinas/farmacología , Ratas , Estereoisomerismo , Relación Estructura-Actividad
6.
J Med Chem ; 55(22): 9416-33, 2012 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-22985112

RESUMEN

There is a high demand for potent, selective, and brain-penetrant small molecule inhibitors of leucine-rich repeat kinase 2 (LRRK2) to test whether inhibition of LRRK2 kinase activity is a potentially viable treatment option for Parkinson's disease patients. Herein we disclose the use of property and structure-based drug design for the optimization of highly ligand efficient aminopyrimidine lead compounds. High throughput in vivo rodent cassette pharmacokinetic studies enabled rapid validation of in vitro-in vivo correlations. Guided by this data, optimal design parameters were established. Effective incorporation of these guidelines into our molecular design process resulted in the discovery of small molecule inhibitors such as GNE-7915 (18) and 19, which possess an ideal balance of LRRK2 cellular potency, broad kinase selectivity, metabolic stability, and brain penetration across multiple species. Advancement of GNE-7915 into rodent and higher species toxicity studies enabled risk assessment for early development.


Asunto(s)
Encéfalo/metabolismo , Morfolinas/farmacología , Enfermedad de Parkinson/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pirimidinas/farmacología , Animales , Diseño de Fármacos , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Macaca fascicularis , Ratones , Ratones Transgénicos , Modelos Moleculares , Morfolinas/síntesis química , Morfolinas/farmacocinética , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/farmacocinética , Pirimidinas/síntesis química , Pirimidinas/farmacocinética , Ratas , Bibliotecas de Moléculas Pequeñas , Distribución Tisular
7.
Toxicol Sci ; 125(2): 496-508, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22094459

RESUMEN

Recent epidemiologic studies have demonstrated a link between organochlorine and pesticide exposure to an enhanced risk for neurodegenerative disorders such as Parkinson's disease (PD). A common biological phenomenon underlying cell injury associated with both polychlorinated biphenyl (PCB) exposure and dopaminergic neurodegeneration during aging is oxidative stress (OS). In this study, we tested the hypothesis that oral PCB exposure, via food ingestion, impairs dopamine systems in the adult murine brain. We determined whether PCB exposure was associated with OS in dopaminergic neurons, a population of cells that selectively degenerate in PD. After 4 weeks of oral exposure to the PCB mixture Aroclor 1254, several congeners, mostly ortho substituted, accumulated throughout the brain. Significant increases in locomotor activity were observed within 2 weeks, which persisted after cessation of PCB exposure. Stereologic analyses revealed a significant loss of dopaminergic neurons within the substantia nigra and ventral tegmental area. However, striatal dopamine levels were elevated, suggesting that compensatory mechanisms exist to maintain dopamine homeostasis, which could contribute to the observed increases in locomotor activity following PCB exposure. Biochemical experiments revealed alterations in OS markers, including increases in SOD and HO-1 levels and the presence of oxidatively modified lipids and proteins. These findings were accompanied by elevated iron levels within the striatal and midbrain regions, perhaps due to the observed dysregulation of transferrin receptors and ferritin levels following PCB exposure. In this study, we suggest that both OS and the uncoupling of iron regulation contribute to dopamine neuron degeneration and hyperactivity following PCB exposure.


Asunto(s)
/toxicidad , Dopamina/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Mesencéfalo/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Administración Oral , Animales , Muerte Celular/efectos de los fármacos , /metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Relación Dosis-Respuesta a Droga , Contaminantes Ambientales/administración & dosificación , Contaminantes Ambientales/metabolismo , Hemo-Oxigenasa 1/metabolismo , Hipercinesia/inducido químicamente , Hipercinesia/metabolismo , Hipercinesia/fisiopatología , Hierro/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Masculino , Proteínas de la Membrana/metabolismo , Mesencéfalo/metabolismo , Mesencéfalo/patología , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Carbonilación Proteica/efectos de los fármacos , Ratas , Superóxido Dismutasa/metabolismo , Factores de Tiempo , Distribución Tisular , Tirosina 3-Monooxigenasa/metabolismo
8.
J Neurosci ; 31(12): 4524-34, 2011 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-21430153

RESUMEN

Dopaminergic neurons of the substantia nigra pars compacta are defective in Parkinson's disease, but the specificity of this dysfunction is not understood. One hypothesis is that mitochondrial bioenergetic capacity is intrinsically lower in striatal dopaminergic presynaptic nerve varicosities, making them unusually susceptible to inhibition of electron transport by oxidative damage. To test this hypothesis, we separated isolated synaptosomes bearing dopamine transporters using immunomagnetic beads and compared their respiration with that of the residual nondopaminergic synaptosomes. As predicted, dopaminergic synaptosomes from striatum had lower respiratory rates. However, so did dopaminergic synaptosomes from cortex, indicating a lack of the predicted striatal specificity. We used fluorescent probes to analyze the bioenergetic competence of individual synaptosomes in the two fractions. The respiratory differences became nonsignificant when respiration rates were normalized to the number of respiration-competent synaptosomes, suggesting that differences reflected the quality of the different fractions. To circumvent damage induced by synaptosomal separation, we monitored membrane potentials in whole unseparated single synaptosomes using fluorescent imaging, and then identified the dopaminergic subpopulation using a fluorescent dopamine transporter substrate (ASP(+) [4-(4-diethylaminostyryl)-N-methylpyridinium iodide]). The capacity of dopaminergic and nondopaminergic synaptosomes to maintain plasma membrane and mitochondrial membrane potential under several stresses did not differ. In addition, this capacity did not decline in either subpopulation with age, a risk factor for Parkinson's disease. We conclude that the intrinsic bioenergetic capacities of dopaminergic and nondopaminergic presynaptic synaptosomes from mice do not differ.


Asunto(s)
Dopamina/fisiología , Metabolismo Energético/fisiología , Estrés Oxidativo/efectos de los fármacos , Sinaptosomas/fisiología , Envejecimiento/fisiología , Animales , Calcio/fisiología , Cuerpo Estriado/fisiología , Dopamina/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Indicadores y Reactivos , Cinética , Masculino , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Mitocondrias/metabolismo , Consumo de Oxígeno , Compuestos de Piridinio/farmacología , Sinaptosomas/metabolismo
9.
J Neurochem ; 112(2): 332-9, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20085612

RESUMEN

The contribution of iron dysregulation to the etiology of a variety of neuronal diseases comes as no surprise given its necessity in numerous general cellular and neuron-specific functions, its abundance, and its highly reactive nature. Homeostatic mechanisms such as the iron regulatory protein and hypoxia-inducible factor pathways are firmly evolutionarily set in place to prevent 'free' iron from participating in chemical Fenton and Haber-Weiss reactions which can result in subsequent generation of toxic hydroxyl radicals. However, given the multiple layers of complexity in cellular iron regulation, disruption of any number of genetic and environmental components can disturb the delicate balance between the various molecular players involved in maintaining an appropriate metabolic iron homeostasis. In this review, we will primarily focus on: (i) the impact of aging and gender on iron dysfunction as these are important criteria in the determination of iron-related disorders such as Parkinson's disease (PD), (ii) how iron mismanagement via disruption of cellular entry and exit pathways may contribute to these disorders, and (iii) how the availability of non-invasive measurement of brain iron may aid in PD diagnosis.


Asunto(s)
Envejecimiento , Encéfalo/metabolismo , Homeostasis/fisiología , Hierro/metabolismo , Enfermedad de Parkinson/patología , Factores de Edad , Animales , Humanos , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/metabolismo , Factores Sexuales
10.
J Biol Chem ; 284(42): 29065-76, 2009 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-19679656

RESUMEN

Hypoxia-inducible factor (HIF) plays an important role in cell survival by regulating iron, antioxidant defense, and mitochondrial function. Pharmacological inhibitors of the iron-dependent enzyme class prolyl hydroxylases (PHD), which target alpha subunits of HIF proteins for degradation, have recently been demonstrated to alleviate neurodegeneration associated with stroke and hypoxic-ischemic injuries. Here we report that inhibition of PHD by 3,4-dihydroxybenzoate (DHB) protects against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced nigral dopaminergic cell loss and up-regulates HIF-1alpha within these neurons. Elevations in mRNA and protein levels of HIF-dependent genes heme oxygenase-1 (Ho-1) and manganese superoxide dismutase (Mnsod) following DHB pretreatment alone are also maintained in the presence of MPTP. MPTP-induced reductions in ferroportin and elevations in nigral and striatal iron levels were reverted to levels comparable with that of untreated controls with DHB pretreatment. Reductions in pyruvate dehydrogenase mRNA and activity resulting from MPTP were also found to be attenuated by DHB. In vitro, the HIF pathway was activated in N27 cells grown at 3% oxygen treated with either PHD inhibitors or an iron chelator. Concordant with our in vivo data, the MPP(+)-elicited increase in total iron as well as decreases in cell viability were attenuated in the presence of DHB. Taken together, these data suggest that protection against MPTP neurotoxicity may be mediated by alterations in iron homeostasis and defense against oxidative stress and mitochondrial dysfunction brought about by cellular HIF-1alpha induction. This study provides novel data extending the possible therapeutic utility of HIF induction to a Parkinson disease model of neurodegeneration, which may prove beneficial not only in this disorder itself but also in other diseases associated with metal-induced oxidative stress.


Asunto(s)
1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/química , Factor 1 Inducible por Hipoxia/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico , Procolágeno-Prolina Dioxigenasa/antagonistas & inhibidores , Animales , Regulación de la Expresión Génica , Hemo-Oxigenasa 1/química , Hidroxibenzoatos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Oxígeno/química , Enfermedad de Parkinson/patología , ARN Mensajero/metabolismo , Ratas
11.
Antioxid Redox Signal ; 11(9): 2083-94, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19290777

RESUMEN

Parkinson's disease (PD) is characterized by early glutathione depletion in the substantia nigra (SN). Among its various functions in the cell, glutathione acts as a substrate for the mitochondrial enzyme glutaredoxin 2 (Grx2). Grx2 is involved in glutathionylation of protein cysteine sulfhydryl residues in the mitochondria. Although monothiol glutathione-dependent oxidoreductases (Grxs) have previously been demonstrated to be involved in iron-sulfur (Fe-S) center biogenesis, including that in yeast, here we report data suggesting the involvement of mitochondrial Grx2, a dithiol Grx, in iron-sulfur biogenesis in a mammalian dopaminergic cell line. Given that mitochondrial dysfunction and increased cellular iron levels are two important hallmarks of PD, this suggests a novel potential mechanism by which glutathione depletion may affect these processes in dopaminergic neurons. We report that depletion of glutathione as substrate results in a dose-dependent Grx2 inhibition and decreased iron incorporation into a mitochondrial complex I (CI) and aconitase (m-aconitase). Mitochondrial Grx2 inhibition through siRNA results in a corresponding decrease in CI and m-aconitase activities. It also results in significant increases in iron-regulatory protein (IRP) binding, likely as a consequence of conversion of Fe-S-containing cellular aconitase to its non-Fe-S-containing IRP1 form. This is accompanied by increased transferrin receptor, decreased ferritin, and subsequent increases in mitochondrial iron levels. This suggests that glutathione depletion may affect important pathologic cellular events associated with PD through its effects on Grx2 activity and mitochondrial Fe-S biogenesis.


Asunto(s)
Dopamina/metabolismo , Glutarredoxinas/metabolismo , Glutatión/metabolismo , Proteínas Hierro-Azufre/metabolismo , Hierro/metabolismo , Mitocondrias/metabolismo , Enfermedad de Parkinson/metabolismo , Animales , Secuencia de Bases , Técnicas de Silenciamiento del Gen , Glutarredoxinas/genética , Espectrometría de Masas , Enfermedad de Parkinson/patología , ARN Interferente Pequeño , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Curr Mol Med ; 6(8): 883-93, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17168739

RESUMEN

A disruption in optimal iron levels within different brain regions has been demonstrated in several neurodegenerative disorders. Although iron is an essential element that is required for many processes in the human body, an excess can lead to the generation of free radicals that can damage cells. Iron levels are therefore stringently regulated within cells by a host of regulatory proteins that keep iron levels in check. The iron regulatory proteins (IRPs) have the ability to sense and control the level of intracellular iron by binding to iron responsive elements (IREs) of several genes encoding key proteins such as the transferrin receptor (TfR) and ferritin. Concurrently, the hypoxia-inducible factor (HIF) has also been shown in previous studies to regulate intracellular iron by binding to HIF-responsive elements (HREs) that are located within the genes of iron-related proteins such as TfR and heme oxygenase-1 (HO-1). This review will focus on the interactions between the IRP/IRE and HIF/HRE systems and how cells utilize these intricate networks to regulate intracellular iron levels. Additionally, since iron chelation has been suggested to be a therapeutic treatment for disorders such as Parkinson's and Alzheimer's disease, understanding the exact mechanisms by which iron acts to cause disease and how the brain would be impacted by iron chelation could potentially give us novel insights into new therapies directed towards preventing or slowing neuronal cell loss associated with these disorders.


Asunto(s)
Inhibidores Enzimáticos/uso terapéutico , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Hierro/metabolismo , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/etiología , Procolágeno-Prolina Dioxigenasa/antagonistas & inhibidores , Regulación de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Quelantes del Hierro/farmacología , Proteínas Reguladoras del Hierro/metabolismo , Elementos de Respuesta/efectos de los fármacos
13.
Mol Interv ; 6(2): 89-97, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16565471

RESUMEN

Iron is essential for many biological processes however excess concentrations can be harmful to many tissues. Its amounts must therefore be carefully regulated in all cells of the body including those in the brain. Increased amounts of iron have been reported in many neurodegenerative disorders. Whether this increased iron contributes to neurodegeneration has been considered controversial. In this review, we discuss some recently identified anomalies in proteins linked with iron metabolism which signify a critical role for iron dysregulation in neurodegeneration.


Asunto(s)
Proteínas Reguladoras del Hierro/metabolismo , Hierro/metabolismo , Degeneración Nerviosa/genética , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/metabolismo , Animales , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Ceruloplasmina/genética , Ceruloplasmina/metabolismo , Hemo-Oxigenasa 1/metabolismo , Proteína de la Hemocromatosis , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Trastornos del Metabolismo del Hierro , Proteínas de Unión a Hierro/genética , Proteínas de Unión a Hierro/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Metaloproteínas/genética , Metaloproteínas/metabolismo , Ratones , Ratones Transgénicos , Modelos Biológicos , Mutación , Enfermedades Neurodegenerativas/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Frataxina
14.
Toxicol Sci ; 90(1): 159-67, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16319092

RESUMEN

Dopaminergic (DAergic) systems have been identified as putative targets for polycholorinated biphenyl (PCB) actions. However, the precise mechanisms leading to neurotoxicity are unresolved. Reactive oxygen species (ROS) were recently shown to mediate injury in DAergic MN9D cells following exposure to Aroclor 1254 (A1254), a commercial PCB mixture. The oxidative stress response in DAergic cells included a persistent expression of heme oxygenase-1 (HO-1). This study tested the hypothesis that a sustained PCB-induced HO-1 response leads to abnormally high Fe levels, which generates ROS production and mediates death in the MN9D DAergic cell model. Accordingly, results indicated that A1254 augmented intracellular Fe levels in MN9D cells after 24 h. Fe chelation by desferoxamine or pharmacologic inhibition of HO activity with tin-protoporphyrin reduced Fe accumulation, ROS production, and cytotoxicity following A1254 exposure. HO-1 over-expression predisposed MN9D DAergic cells to enhanced ROS production and cell death in response to PCBs. Conversely, antisense inhibition of HO-1 expression prevented PCB-induced ROS production and cell death. These observations suggest that enhanced HO-1 catalytic activity and subsequent liberation of Fe participate in neurotoxic DAergic cell injury caused by A1254 exposure in vitro.


Asunto(s)
/toxicidad , Dopamina/metabolismo , Contaminantes Ambientales/toxicidad , Hemo-Oxigenasa 1/metabolismo , Neuronas/efectos de los fármacos , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Deferoxamina/farmacología , Inhibidores Enzimáticos/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hemo-Oxigenasa 1/antagonistas & inhibidores , Hemo-Oxigenasa 1/genética , Hierro/metabolismo , Metaloporfirinas/farmacología , Ratones , Neuronas/metabolismo , Oligorribonucleótidos Antisentido/farmacología , Estrés Oxidativo/efectos de los fármacos , Protoporfirinas/farmacología , Especies Reactivas de Oxígeno/metabolismo
15.
Neurotoxicology ; 26(1): 63-75, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15527874

RESUMEN

A lack of evidence supporting a role of heritability in the development of idiopathic Parkinson's disease (PD) has implicated exposures to environmental contaminants in the disease etiology. Epidemiological and clinical studies, as well as animal models of the PD phenotype, have consistently linked agrichemical exposure with dopaminergic (DAergic) damage, particularly through oxidative stress mechanisms. Maneb (MB) is a dithiocarbamate (DTC) fungicide that has specifically been implicated to have adverse effects on dopamine (DA) systems, but the role MB plays in modulating the oxidative state of DAergic cells has not previously been described. Since glutathione (GSH) is a major cellular antioxidant, it was hypothesized that exposure to MB would disrupt this system. The current study primarily utilized the PC12 cell line, which displays a catecholaminergic phenotype. Low concentrations of MB (50-1000 ng/ml) had little effect on cell viability, as measured by LDH release. These same concentrations, however, led to increases in GSH and its oxidized form, GSSG. Effects on viability and GSH were correlated to a primary mesencephalic culture system. Furthermore, these effects were markedly different from those observed with the classical oxidative stressor and pesticide, paraquat (PQ). To determine how MB would affect cells in which antioxidant systems were compromised, PC12 cells were treated with L-buthionine-(S,R)-sulfoximine (BSO) to deplete cellular GSH, followed by treatment with MB. Results suggest that following an insult to the GSH antioxidant system, MB can act as an additional insult to the system and prevent the normal recovery of those defenses. Altered protein levels of heme oxygenase-1 (HO-1) further indicated an oxidative stress response elicited by MB in PC12 cells. DAergic neurons, as a population, are inherently vulnerable to oxidative stress, and the disruption of antioxidant systems by the fungicide MB may contribute to the neurodegeneration of these cells, especially with concurrent exposures to other environmentally relevant oxidative stressors, such as PQ.


Asunto(s)
Antioxidantes/metabolismo , Dopamina/fisiología , Fungicidas Industriales/toxicidad , Maneb/toxicidad , Actinas/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Glutamato-Cisteína Ligasa/metabolismo , Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemo-Oxigenasa 1 , Immunoblotting , L-Lactato Deshidrogenasa/metabolismo , Mesencéfalo/citología , Mesencéfalo/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas
16.
Mol Cell Proteomics ; 3(10): 950-9, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15220401

RESUMEN

Human adenovirus proteinase (AVP) requires two cofactors for maximal activity: pVIc, a peptide derived from the C terminus of adenovirus precursor protein pVI, and the viral DNA. Synchrotron protein footprinting was used to map the solvent accessible cofactor binding sites and to identify conformational changes associated with the binding of cofactors to AVP. The binding of pVIc alone or pVIc and DNA together to AVP triggered significant conformational changes adjacent to the active site cleft sandwiched between the two AVP subdomains. In addition, upon binding of DNA to AVP, it was observed that specific residues on each of the two major subdomains were significantly protected from hydroxyl radicals. Based on the locations of these protected side-chain residues and conserved aromatic and positively charged residues within AVP, a three-dimensional model of DNA binding was constructed. The model indicated that DNA binding can alter the relative orientation of the two AVP domains leading to the partial activation of AVP by DNA. In addition, both pVIc and DNA may independently alter the active site conformation as well as drive it cooperatively to fully activate AVP.


Asunto(s)
Adenovirus Humanos/enzimología , ADN Viral/metabolismo , Endopeptidasas/metabolismo , Activación Enzimática , Proteínas Virales/metabolismo , Adenovirus Humanos/genética , Secuencia de Aminoácidos , Aminoácidos Acídicos , Aminoácidos Básicos , Sitios de Unión , Endopeptidasas/química , Humanos , Modelos Moleculares , Conformación Proteica , Huella de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Sincrotrones , Proteínas Virales/química , Proteínas Virales/genética
17.
J Pharmacol Exp Ther ; 305(3): 872-9, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12626652

RESUMEN

We have previously reported that CYP3A cross-links with polyubiquitinated proteins in microsomes from nicardipine-treated rats in a process that is distinct from classical polyubiquitination. To further examine the role of the proteasome in CYP3A degradation, we investigated the effects of proteasome inhibitors lactacystin, MG132, proteasome inhibitor 1, and hemin in primary cultures of rat and human hepatocytes. With the exception of hemin, these agents increased the total pool of ubiquitinated proteins in microsomes isolated from rat hepatocytes, indicating that lactacystin, MG132, and proteasome inhibitor 1 effectively inhibited the proteasome in these cells. All four agents caused a reduction in the amount of the major approximately 55-kDa CYP3A band, opposite to what would be expected if the ubiquitin-proteasome pathway degraded CYP3A. Only hemin treatment caused an increase in high molecular mass (HMM) CYP3A bands. Because hemin treatment did not alter levels of ubiquitin in CYP3A immunoprecipitates, the HMM CYP3A bands formed in response to hemin treatment clearly were not due to proteasome inhibition. Rather, because hemin treatment also caused an increase in HMM CYP3A in the detergent-insoluble fraction of the 10,000g pellet, the HMM CYP3A seems to represent a large protein complex that is unlikely to primarily represent ubiquitination.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/metabolismo , Hepatocitos/efectos de los fármacos , Leupeptinas/farmacología , Complejos Multienzimáticos/antagonistas & inhibidores , Oxidorreductasas N-Desmetilantes/metabolismo , Animales , Cisteína Endopeptidasas , Inhibidores de Cisteína Proteinasa/farmacología , Citocromo P-450 CYP3A , Silenciador del Gen/efectos de los fármacos , Hepatocitos/enzimología , Humanos , Complejo de la Endopetidasa Proteasomal , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA